1
|
Joldes C, Jimbu L, Mesaros O, Zdrenghea M, Fetica B. Tumor-Associated Macrophages as Key Modulators of Disease Progression in Diffuse Large B-Cell Lymphoma. Biomedicines 2025; 13:1099. [PMID: 40426926 PMCID: PMC12108958 DOI: 10.3390/biomedicines13051099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/17/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025] Open
Abstract
With the advent of new therapeutic approaches, there is hope that anticancer treatment will eventually be possible without the use of chemotherapy. Efficient immunotherapeutic options have recently emerged in many cancers, offering a less aggressive approach, with overall better tolerance, making them also suitable for frail patients. Response to immunotherapy relies on the availability, functionality, and efficacy of the host's immune effector mechanisms. One of the key factors determining the efficacy of immunotherapy is the tumor microenvironment, which encompasses various immune effectors, including macrophages, which play a crucial role in regulating immune responses through phagocytosis and antigen presentation. Macrophages are prototypically divided, according to their polarization, into either the pro-inflammatory M1 type or the anti-inflammatory M2 type. In the tumor microenvironment, M2-polarized macrophages, known as tumor-associated macrophages (TAMs), are the predominant phenotype and are associated with tumor progression. The M1/M2 paradigm contributes to the understanding of tumor progression. Due to the variable microenvironment, the mechanisms regulating TAMs can vary across different cancers. Variations in TAM polarization may account for the different treatment responses in patients with similar diseases. This paper investigates the connection between TAMs, disease progression, and treatment responses in the most frequent solid hematologic cancer, diffuse large B-cell lymphoma.
Collapse
Affiliation(s)
- Corina Joldes
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
| | - Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Bogdan Fetica
- Department of Pathology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania;
| |
Collapse
|
2
|
Saris J, Li Yim AYF, Bootsma S, Lenos KJ, Franco Fernandez R, Khan HN, Verhoeff J, Poel D, Mrzlikar NM, Xiong L, Schijven MP, van Grieken NCT, Kranenburg O, Wildenberg ME, Logiantara A, Jongerius C, Garcia Vallejo JJ, Gisbertz SS, Derks S, Tuynman JB, D'Haens GRAM, Vermeulen L, Grootjans J. Peritoneal resident macrophages constitute an immunosuppressive environment in peritoneal metastasized colorectal cancer. Nat Commun 2025; 16:3669. [PMID: 40246872 PMCID: PMC12006467 DOI: 10.1038/s41467-025-58999-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Patients with peritoneal metastasized colorectal cancer (PM-CRC) have a dismal prognosis. We hypothesized that an immunosuppressive environment in the peritoneal cavity underlies poor prognosis. We define the composition of the human peritoneal immune system (PerIS) using single-cell technologies in 18 patients with- and without PM-CRC, as well as in matched peritoneal metastases (n = 8). Here we show that the PerIS contains abundant immunosuppressive C1Q+VSIG4+ and SPP1+VSIG4+ peritoneal-resident macrophages (PRMs), as well as monocyte-like cavity macrophages (mono-CMs), which share features with tumor-associated macrophages, even in homeostasis. In PM-CRC, expression of immunosuppressive cytokines IL10 and VEGF increases, while simultaneously expression of antigen-presenting molecules decreases in PRMs. These intratumoral suppressive PRMs originate from the PerIS, and intraperitoneal depletion of PRMs in vivo using anti-CSF1R combined with anti-PD1 significantly reduces tumor burden and improves survival. Thus, PRMs define a metastatic site-specific immunosuppressive niche, and targeting PRMs is a promising treatment strategy for PM-CRC.
Collapse
Affiliation(s)
- J Saris
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - A Y F Li Yim
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands
| | - S Bootsma
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - K J Lenos
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - R Franco Fernandez
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - H N Khan
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - J Verhoeff
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands
- Molecular Cell Biology & Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - D Poel
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - N M Mrzlikar
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - L Xiong
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - M P Schijven
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Digital Health, Amsterdam, The Netherlands
| | - N C T van Grieken
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - O Kranenburg
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, The Netherlands
- Utrecht Platform for Organoid Technology, Utrecht University, Utrecht, The Netherlands
| | - M E Wildenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A Logiantara
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - C Jongerius
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - J J Garcia Vallejo
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands
- Molecular Cell Biology & Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - S S Gisbertz
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - S Derks
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J B Tuynman
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - G R A M D'Haens
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - L Vermeulen
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
- Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - J Grootjans
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Amsterdam, The Netherlands.
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Oncode Institute, Amsterdam, The Netherlands.
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Xie G, Jiang G, Huang L, Sun S, Li X, Wu B, Wang H, Zhang Z, Ye K, Yu Y, Xiong J. Asparagine Endopeptidase Inhibition Attenuates Tissue Plasminogen Activator-Induced Brain Hemorrhagic Transformation After Ischemic Stroke. CNS Neurosci Ther 2025; 31:e70345. [PMID: 40116141 PMCID: PMC11926568 DOI: 10.1111/cns.70345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Thrombolytic treatment with tissue plasminogen activator (tPA) is one of the approved pharmacological therapies for acute ischemic stroke. However, the use of tPA is limited due to hemorrhagic transformation (HT) and the narrow therapeutic time window. Previous studies demonstrated that asparagine endopeptidase (AEP), a widely expressed pH-dependent endo-lysosomal cysteine protease, can induce neuronal death during ischemia-reperfusion injury. But whether AEP is engaged in HT during ischemia-reperfusion injury is unclear. In the current study, we expanded the role of AEP on HT after delayed tPA administration. METHODS In order to investigate the effects of AEP on HT after delayed tPA administration following ischemic stroke, the middle cerebral artery occlusion/reperfusion (MCAO/R) was performed in wild-type (WT) and AEP knockout (KO) transgenic mice, followed by delayed administration of tPA (10 mg/kg, 3 h after occlusion). Additionally, we explored the potential of R13, a specific TrkB agonist with a strong inhibitory impact on AEP, to mitigate injury induced by tPA. 24 h after tPA administration, the following parameters were assessed: infarct volume, behavioral tests, hemorrhagic levels, Evans blue leakage, tight and adherens junction protein expression, blood-brain barrier (BBB) function, cerebral vascular structure, matrix metalloproteinases (MMPs), and BBB-regulated protein low-density lipoprotein receptor-related protein 1 (LRP-1) expression. To construct an in vitro model to examine the effects of AEP on ischemia-reperfusion injury after tPA treatment, human umbilical vein endothelial cells (HUVECs) were exposed to 4 h of oxygen-glucose deprivation (OGD), followed by treatment with tPA (500 ng/mL). 7,8-dihydroxyflavone (7,8-DHF), a natural TrkB agonist with an inhibitory effect on AEP, was applied before OGD. RESULTS Compared with tPA-treated WT mice, AEP KO mice treated with tPA showed improved infarct volume, neurological function, brain edema, brain hemoglobin levels, Evans blue leakage, vascular tight junctions, and basement membrane structure combined with reduced AEP expression and activity within the peri-infarct area. In addition, the mice treated with R13 exhibited protective effects on the BBB. Furthermore, we found that the expression of MMP2, MMP9, and LRP-1 in the brain was inhibited by both AEP knockout and R13 treatment. Moreover, HUVECs treated with 7,8-DHF showed improvements in tight and adherens junction proteins and suppressed levels of MMP2, MMP9, and LRP-1. CONCLUSION Our findings demonstrate that AEP exacerbates HT induced by delayed tPA treatment in acute ischemic stroke by activating LRP-1, MMP2, and MMP9, which disrupts BBB integrity. We further confirmed R13 as a preventive therapy to attenuate HT induced by delayed tPA treatment in acute ischemic stroke. The present study suggests AEP inhibition may serve as a promising strategy to enhance the safety of delayed tPA thrombolysis for ischemic stroke.
Collapse
Affiliation(s)
- Guanfeng Xie
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - Gege Jiang
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - Liqin Huang
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - Shangqi Sun
- Department of Neurology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Xiaoyi Li
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - Bingjie Wu
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - Hualong Wang
- Department of NeurologyThe First Hospital of Hebei Medical University, Brain Aging and Cognitive Neuroscience Laboratory of Hebei ProvinceShijiazhuangHebeiChina
| | - Zhentao Zhang
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdong ProvinceChina
| | - Ying Yu
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - Jing Xiong
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
- Taikang Center for Life and Medical SciencesWuhan UniversityWuhanChina
| |
Collapse
|
4
|
Lustenberger SK, Castro Jaramillo CA, Bärtschi LA, Williams R, Schibli R, Mu L, Krämer SD. Towards imaging the immune state of cancer by PET: Targeting legumain with 11C-labeled P1-Asn peptidomimetics carrying a cyano-warhead. Nucl Med Biol 2024; 138-139:108951. [PMID: 39303441 DOI: 10.1016/j.nucmedbio.2024.108951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE M2-type tumor-associated macrophages (TAM) residing in the tumor microenvironment (TME) have been linked to tumor invasiveness, metastasis and poor prognosis. M2 TAMs suppress T cell activation, silencing the recognition of the cancer by the immune system. Targeting TAMs in anti-cancer therapy may support the immune system and immune-checkpoint inhibitor therapies to fight the cancer cells. We aimed to develop a PET tracer for the imaging of M2 TAM infiltration of cancer, using activated legumain as the imaging target. BASIC PROCEDURES Two P1-mimicking inhibitors with a cyano-warhead were labeled with carbon-11 and evaluated in vitro and in vivo with a CT26 tumor mouse model. Target expression and activity were quantified from RT-qPCR and in vitro substrate conversion, respectively. The co-localization of legumain and TAMs was assessed by fluorescence microscopy. The two tracers were evaluated by PET with subsequent biodistribution analysis with the dissected tissues. Parent-to-total radioactivity in plasma was determined at several time points after i.v. tracer injection, using reverse phase radio-UPLC. MAIN FINDINGS Legumain displayed a target density of 40.7 ± 19.1 pmol per mg total protein in tumor lysate (n = 4) with high substrate conversion and colocalization with M2 macrophages in the tumor periphery. [11C]1 and [11C]2 were synthesized with >95 % radiochemical purity and 12.9-382.2 GBq/μmol molar activity at the end of synthesis. We observed heterogeneous tumor accumulation in in vitro autoradiography and PET for both tracers. However, excess unlabeled 1 or 2 did not compete with tracer accumulation. Both [11C]1 and [11C]2 were rapidly metabolized to a polar radiometabolite in vivo. PRINCIPAL CONCLUSIONS The legumain tracers [11C]1 and [11C]2, synthesized with high radiochemical purity and molar activity, accumulate in the legumain-positive CT26 tumor in vivo. However, the lack of competition by excess compound questions their specificity. Both tracers are rapidly metabolized in vivo, requiring structural modifications towards more stable tracers for further investigations.
Collapse
Affiliation(s)
- Severin K Lustenberger
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Claudia A Castro Jaramillo
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Lena A Bärtschi
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Rich Williams
- Queens University Belfast, BT7 1NN Belfast, United Kingdom of Great Britain and Northern Ireland
| | - Roger Schibli
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Linjing Mu
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Stefanie D Krämer
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland.
| |
Collapse
|
5
|
Cao Z, Leng P, Xu H, Li X. The regulating role of galectin-9 in immune cell populations. Front Pharmacol 2024; 15:1462061. [PMID: 39539619 PMCID: PMC11557436 DOI: 10.3389/fphar.2024.1462061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Galectin-9 (gal-9) is a protein that belongs to the galectin family. Gal-9 is expressed in cells of the innate and adaptive immune system, including lymphocytes, dendritic cells, giant salivary cells, eosinophils and T cells, etc. In different immune cells, the role of gal-9 is different. Gal-9 can induce the proliferation and activation of immune cells, and also promote the apoptosis of immune cells. This effect of gal-9 affects the occurrence and development of a variety of immune-related diseases, such as the invasion of pathogenic microorganisms, immune escape of tumor cells, and inflammatory response. Thus, understanding the biological roles of gal-9 in innate and adaptive immunity may be essential for autoimmune diseases treatment and diagnosis to improve patient quality of life. In this review, we aim to summarize current research on the regulatory roles of gal-9 in human immune system and potential inducers and inhibitors of gal-9, which may provide new strategies for immune diseases therapies.
Collapse
Affiliation(s)
- Zhanqi Cao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | | | | |
Collapse
|
6
|
Zhou Y, Lou J, Tian Y, Ding J, Wang X, Tang B. How lactate affects immune strategies in lymphoma. Front Mol Biosci 2024; 11:1480884. [PMID: 39464313 PMCID: PMC11502318 DOI: 10.3389/fmolb.2024.1480884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
Tumor cells undergo metabolic reprogramming through shared pathways, resulting in a hypoxic, acidic, and highly permeable internal tumor microenvironment (TME). Lactate, once only regarded as a waste product of glycolysis, has an inseparable dual role with tumor immunity. It can not only provide a carbon source for immune cells to enhance immunity but also help the immune escape through a variety of ways. Lymphoma also depends on the proliferation signal of TME. This review focuses on the dynamic process of lactate metabolism and immune function changes in lymphoma and aims to comprehensively summarize and explore which genes, transcription factors, and pathways affect the biological changes and functions of immune cells. To deeply understand the complex and multifaceted role of lactate metabolism and immunity in lymphoma, the combination of lactate targeted therapy and classical immunotherapy will be a promising development direction in the future.
Collapse
Affiliation(s)
- Yuehan Zhou
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinzhan Lou
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuqin Tian
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinlei Ding
- Department of Thoracic Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaobo Wang
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bo Tang
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Forbord KM, Lunde NN, Bosnjak-Olsen T, Johansen HT, Solberg R, Jafari A. Legumain is a paracrine regulator of osteoblast differentiation and mediates the inhibitory effect of TGF-β1 on osteoblast maturation. Front Endocrinol (Lausanne) 2024; 15:1445049. [PMID: 39363898 PMCID: PMC11446771 DOI: 10.3389/fendo.2024.1445049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) is a critical regulator of skeletal homeostasis and has diverse effects on osteoblastogenesis. To date, the mechanisms behind the intriguing inhibitory effect of TGF-β1 on osteoblast maturation are not fully understood. Here, we demonstrate a novel mechanism by which TGF-β1 modulates osteoblast maturation through the lysosomal protease legumain. We observed that addition of TGF-β1 to osteogenic cultures of human bone marrow derived mesenchymal stromal (stem) cells enhanced legumain activity and secretion, in-spite of decreased legumain mRNA expression, suggesting post-transcriptional regulation. We further showed that osteogenic cells internalize and activate prolegumain, associated with inhibited osteoblast maturation, revealing legumain as a paracrine regulator of osteoblast maturation. Interestingly, TGF-β1 treatment exacerbated legumain internalization and activity, and showed an additive effect on legumain-induced inhibition of osteoblast maturation. Importantly, pharmacological inhibition of legumain abolished the inhibitory effect of TGF-β1 on osteoblast maturation. Our findings reveal that TGF-β1 inhibits osteoblast maturation by stimulating secretion and activity of endogenous legumain, as well as enhancing internalization and activation of extracellular prolegumain. Therefore, our study provides a deeper understanding of the complex regulation of osteoblastogenesis and unveils a novel TGF-β1-legumain axis in regulation of osteoblast maturation, offering novel insights for possible therapeutic interventions related to bone diseases associated with aberrant TGF-β1 signaling.
Collapse
Affiliation(s)
- Karl Martin Forbord
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Ngoc Nguyen Lunde
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Tatjana Bosnjak-Olsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Harald Thidemann Johansen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Rigmor Solberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Gray ME, Zielinski KM, Xu F, Elder KK, McKay SJ, Ojo VT, Benjamin SR, Yaseen AA, Brooks TA, Tumey LN. A comparison of the activity, lysosomal stability, and efficacy of legumain-cleavable and cathepsin-cleavable ADC linkers. Xenobiotica 2024; 54:458-468. [PMID: 38738708 PMCID: PMC11436314 DOI: 10.1080/00498254.2024.2352051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024]
Abstract
Over the past two decades, antibody-drug conjugates (ADCs) have emerged as a highly effective drug delivery technology. ADCs utilise a monoclonal antibody, a chemical linker, and a therapeutic payload to selectively deliver highly potent pharmaceutical agents to specific cell types.Challenges such as premature linker cleavage and clearance due to linker hydrophobicity have adversely impacted the stability and safety of ADCs. While there are various solutions to these challenges, our team has focused on replacement of hydrophobic ValCit linkers (cleaved by CatB) with Asn-containing linkers that are cleaved by lysosomal legumain.Legumain is abundantly present in lysosomes and is known to play a role in tumour microenvironment dynamics. Herein, we directly compare the lysosomal cleavage, cytotoxicity, plasma stability, and efficacy of a traditional cathepsin-cleavable ADC to a matched Asn-containing legumain-cleavable ADC.We demonstrate that Asn-containing linker sequences are specifically cleaved by lysosomal legumain and that Asn-linked MMAE ADCs are broadly active against a variety of tumours, even those with low legumain expression. Finally, we show that AsnAsn-linked ADCs exhibit comparable or improved efficacy to traditional ValCit-linked ADCs. Our study paves the way for replacement of the traditional ValCit linker technology with more hydrophilic Asn-containing peptide linker sequences.
Collapse
Affiliation(s)
- Meghan E Gray
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Karina M Zielinski
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Fanny Xu
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Kayla K Elder
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Steven J McKay
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Victor T Ojo
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Samantha R Benjamin
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Aiman A Yaseen
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Tracy A Brooks
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - L Nathan Tumey
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| |
Collapse
|
9
|
Li K, Nie H, Jin R, Wu X. Mesenchymal stem cells-macrophages crosstalk and myeloid malignancy. Front Immunol 2024; 15:1397005. [PMID: 38779660 PMCID: PMC11109455 DOI: 10.3389/fimmu.2024.1397005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
As major components of the tumor microenvironment, both mesenchymal stem cells (MSCs) and macrophages can be remodelled and exhibit different phenotypes and functions during tumor initiation and progression. In recent years, increasing evidence has shown that tumor-associated macrophages (TAMs) play a crucial role in the growth, metastasis, and chemotherapy resistance of hematological malignancies, and are associated with poor prognosis. Consequently, TAMs have emerged as promising therapeutic targets. Notably, MSCs exert a profound influence on modulating immune cell functions such as macrophages and granulocytes, thereby playing a crucial role in shaping the immunosuppressive microenvironment surrounding tumors. However, in hematological malignancies, the cellular and molecular mechanisms underlying the interaction between MSCs and macrophages have not been clearly elucidated. In this review, we provide an overview of the role of TAMs in various common hematological malignancies, and discuss the latest advances in understanding the interaction between MSCs and macrophages in disease progression. Additionally, potential therapeutic approaches targeting this relationship are outlined.
Collapse
Affiliation(s)
- Kun Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Nie
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Koudouna A, Gkioka AI, Gkiokas A, Tryfou TM, Papadatou M, Alexandropoulos A, Bartzi V, Kafasi N, Kyrtsonis MC. Serum-Soluble CD163 Levels as a Prognostic Biomarker in Patients with Diffuse Large B-Cell Lymphoma Treated with Chemoimmunotherapy. Int J Mol Sci 2024; 25:2862. [PMID: 38474108 DOI: 10.3390/ijms25052862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The majority of patients with Diffuse Large B-cell Lymphoma (DLBCL) will respond to first-line treatment and be cured. However, the disease is heterogeneous, and biomarkers able to discriminate patients with suboptimal prognosis are needed. M2 CD163-positive tumor-associated macrophages (TAMs) were shown to be implicated in DLBCL disease activity regulation. Serum-soluble CD163 (sCD163) functions as a scavenger receptor for haptoglobin-hemoglobin complexes and is mostly expressed by monocytes and macrophages. Its levels are used to determine macrophage activation. We aimed to determine serum sCD163 in a sample of DLBCL patients and study eventual correlations with parameters of disease activity or survival. Serum sCD163 levels were measured in 40 frozen sera from patients diagnosed with DLBCL and 30 healthy individuals (HIs) using an enzyme-linked immunosorbent assay (ELISA). Statistical analyses were performed using SPSS version 28. The results showed that patients who achieved complete response after standard-of-care immunochemotherapy and were alive and disease-free after 12 months of follow-up but had elevated sCD163 levels (above median) at diagnosis presented a significantly worse overall survival compared to those with initial serum sCD163 levels below the median (p = 0.03). Consequently, serum sCD163 levels in patients with DLBCL may constitute a marker of long-term response to chemoimmunotherapy.
Collapse
Affiliation(s)
- Aspasia Koudouna
- Hematology Section, First Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens' Medical School, 11527 Athens, Greece
| | - Annita Ioanna Gkioka
- Hematology Section, First Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens' Medical School, 11527 Athens, Greece
| | - Alexandros Gkiokas
- Hematology Section, First Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens' Medical School, 11527 Athens, Greece
| | - Thomai M Tryfou
- Hematology Section, First Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens' Medical School, 11527 Athens, Greece
| | - Mavra Papadatou
- Hematology Section, First Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens' Medical School, 11527 Athens, Greece
| | - Alexandros Alexandropoulos
- Hematology Section, First Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens' Medical School, 11527 Athens, Greece
| | - Vassiliki Bartzi
- Hematology Section, First Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens' Medical School, 11527 Athens, Greece
| | | | - Marie-Christine Kyrtsonis
- Hematology Section, First Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian University of Athens' Medical School, 11527 Athens, Greece
| |
Collapse
|
11
|
Cai F, Zhang J, Gao H, Shen H. Tumor microenvironment and CAR-T cell immunotherapy in B-cell lymphoma. Eur J Haematol 2024; 112:223-235. [PMID: 37706523 DOI: 10.1111/ejh.14103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Chimeric receptor antigen T cell (CAR-T cell) therapy has demonstrated effectiveness and therapeutic potential in the immunotherapy of hematological malignancies, representing a promising breakthrough in cancer treatment. Despite the efficacy of CAR-T cell therapy in B-cell lymphoma, response variability, resistance, and side effects remain persistent challenges. The tumor microenvironment (TME) plays an intricate role in CAR-T cell therapy of B-cell lymphoma. The TME is a complex and dynamic environment that includes various cell types, cytokines, and extracellular matrix components, all of which can influence CAR-T cell function and behavior. This review discusses the design principles of CAR-T cells, TME in B-cell lymphoma, and the mechanisms by which TME influences CAR-T cell function. We discuss emerging strategies aimed at modulating the TME, targeting immunosuppressive cells, overcoming inhibitory signaling, and improving CAR-T cell infiltration and persistence. Therefore, these processes enhance the efficacy of CAR-T cell therapy and improve patient outcomes in B-cell lymphoma. Further research will be needed to investigate the molecular and cellular events that occur post-infusion, including changes in TME composition, immune cell interactions, cytokine signaling, and potential resistance mechanisms. Understanding these processes will contribute to the development of more effective CAR-T cell therapies and strategies to mitigate treatment-related toxicities.
Collapse
Affiliation(s)
- Fengqing Cai
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Junfeng Zhang
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Hui Gao
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Hongqiang Shen
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Department of Hematology-Oncology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Joint Research Center for Immune Landscape and Precision Medicine in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Ribatti D, Tamma R, Annese T, Ingravallo G, Specchia G. Macrophages and angiogenesis in human lymphomas. Clin Exp Med 2024; 24:26. [PMID: 38285283 PMCID: PMC10824884 DOI: 10.1007/s10238-023-01291-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 12/27/2023] [Indexed: 01/30/2024]
Abstract
A link exists between chronic inflammation and cancer and immune cells, angiogenesis, and tumor progression. In hematologic malignancies, tumor-associated macrophages (TAMs) are a significant part of the tumor microenvironment. Macrophages are classified into M1/classically activated and M2/alternatively activated. In tumors, TAMs are mainly constituted by M2 subtype, which promotes angiogenesis, lymphangiogenesis, repair, and remodeling, suppressing adaptive immunity, increasing tumor cell proliferation, drug resistance, histological malignancy, and poor clinical prognosis. The aim of our review article is to define the role of TAMs and their relationship with the angiogenesis in patients with lymphoma reporting both an analysis of main published data and those emerging from our studies. Finally, we have discussed the anti-angiogenic approach in the treatment of lymphomas.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy.
| | - Roberto Tamma
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy
| | - Tiziana Annese
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy
- Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, Bari, Italy
| | - Giuseppe Ingravallo
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Medical School, Bari, Italy
| | | |
Collapse
|
13
|
Xiong J, Xiao R, Zhao J, Zhao Q, Luo M, Li F, Zhang W, Wu M. Matrix stiffness affects tumor-associated macrophage functional polarization and its potential in tumor therapy. J Transl Med 2024; 22:85. [PMID: 38246995 PMCID: PMC10800063 DOI: 10.1186/s12967-023-04810-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/17/2023] [Indexed: 01/23/2024] Open
Abstract
The extracellular matrix (ECM) plays critical roles in cytoskeletal support, biomechanical transduction and biochemical signal transformation. Tumor-associated macrophage (TAM) function is regulated by matrix stiffness in solid tumors and is often associated with poor prognosis. ECM stiffness-induced mechanical cues can activate cell membrane mechanoreceptors and corresponding mechanotransducers in the cytoplasm, modulating the phenotype of TAMs. Currently, tuning TAM polarization through matrix stiffness-induced mechanical stimulation has received increasing attention, whereas its effect on TAM fate has rarely been summarized. A better understanding of the relationship between matrix stiffness and macrophage function will contribute to the development of new strategies for cancer therapy. In this review, we first introduced the overall relationship between macrophage polarization and matrix stiffness, analyzed the changes in mechanoreceptors and mechanotransducers mediated by matrix stiffness on macrophage function and tumor progression, and finally summarized the effects of targeting ECM stiffness on tumor prognosis to provide insight into this new field.
Collapse
Affiliation(s)
- Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Rourou Xiao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jiahui Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qiuyan Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Manwen Luo
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Feng Li
- Department of Medical Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan, 430071, China.
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, China.
| |
Collapse
|
14
|
Xiong D, Wei X, Huang W, Zheng J, Feng R. Prediction significance of autophagy-related genes in survival probability and drug resistance in diffuse large B-cell lymphoma. Aging (Albany NY) 2024; 16:1049-1076. [PMID: 38240686 PMCID: PMC10866451 DOI: 10.18632/aging.205282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/15/2023] [Indexed: 02/06/2024]
Abstract
BACKGROUND/AIMS Diffuse large B-cell lymphoma (DLBCL), the most common subtype of non-Hodgkin lymphoma, has significant prognostic heterogeneity. This study aimed to generate a prognostic prediction model based on autophagy-related genes for DLBCL patients. METHODS Utilizing bioinformatics techniques, we analyzed the clinical information and transcriptome data of DLBCL patients from the Gene Expression Omnibus (GEO) database. Through unsupervised clustering, we identified new autophagy-related molecular subtypes and pinpointed differentially expressed genes (DEGs) between these subtypes. Based on these DEGs, a prognostic model was constructed using Cox and Lasso regression. The effectiveness, accuracy, and clinical utility of this prognostic model were assessed using numerous independent validation cohorts, survival analyses, receiver operating characteristic (ROC) curves, multivariate Cox regression analysis, nomograms, and calibration curves. Moreover, functional analysis, immune cell infiltration, and drug sensitivity analysis were performed. RESULTS DLBCL patients with different clinical characterizations (age, molecular subtypes, ECOG scores, and stages) showed different expression features of autophagy-related genes. The prediction model was constructed based on the eight autophagy-related genes (ADD3, IGFBP3, TPM1, LYZ, AFDN, DNAJC10, GLIS3, and CCDC102A). The prognostic nomogram for overall survival of DLBCL patients incorporated risk level, stage, ECOG scores, and molecular subtypes, showing excellent agreement between observed and predicted outcomes. Differences were noted in the proportions of immune cells (native B cells, Treg cells, CD8+ T cell, CD4+ memory activated T cells, gamma delta T cells, macrophages M1, and resting mast cells) between high-risk and low-risk groups. LYZ and ADD3 exhibited correlations with drug resistance to most chemotherapeutic drugs. CONCLUSIONS This study established a novel prognostic assessment model based on the expression profile of autophagy-related genes and clinical characteristics of DLBCL patients, explored immune infiltration and predicted drug resistance, which may guide precise and individualized immunochemotherapy regimens.
Collapse
Affiliation(s)
- Dan Xiong
- Department of Hematology, Nanfang Hospital, Southern Medical University or the First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- Department of Hematology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan 528308, Guangdong, China
| | - Xiaolei Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University or the First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Weiming Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University or the First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jingxia Zheng
- Department of Hematology, Nanfang Hospital, Southern Medical University or the First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ru Feng
- Department of Hematology, Nanfang Hospital, Southern Medical University or the First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
15
|
Bandi DR, Chitturi CMK, Aswathanarayan JB, Veeresh PKM, Bovilla VR, Sukocheva OA, Devi PS, Natraj SM, Madhunapantula SV. Pigmented Microbial Extract (PMB) from Exiguobacterium Species MB2 Strain (PMB1) and Bacillus subtilis Strain MB1 (PMB2) Inhibited Breast Cancer Cells Growth In Vivo and In Vitro. Int J Mol Sci 2023; 24:17412. [PMID: 38139241 PMCID: PMC10743659 DOI: 10.3390/ijms242417412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Breast cancer (BC) continues to be one of the major causes of cancer deaths in women. Progress has been made in targeting hormone and growth factor receptor-positive BCs with clinical efficacy and success. However, little progress has been made to develop a clinically viable treatment for the triple-negative BC cases (TNBCs). The current study aims to identify potent agents that can target TNBCs. Extracts from microbial sources have been reported to contain pharmacological agents that can selectively inhibit cancer cell growth. We have screened and identified pigmented microbial extracts (PMBs) that can inhibit BC cell proliferation by targeting legumain (LGMN). LGMN is an oncogenic protein expressed not only in malignant cells but also in tumor microenvironment cells, including tumor-associated macrophages. An LGMN inhibition assay was performed, and microbial extracts were evaluated for in vitro anticancer activity in BC cell lines, angiogenesis assay with chick chorioallantoic membrane (CAM), and tumor xenograft models in Swiss albino mice. We have identified that PMB from the Exiguobacterium (PMB1), inhibits BC growth more potently than PMB2, from the Bacillus subtilis strain. The analysis of PMB1 by GC-MS showed the presence of a variety of fatty acids and fatty-acid derivatives, small molecule phenolics, and aldehydes. PMB1 inhibited the activity of oncogenic legumain in BC cells and induced cell cycle arrest and apoptosis. PMB1 reduced the angiogenesis and inhibited BC cell migration. In mice, intraperitoneal administration of PMB1 retarded the growth of xenografted Ehrlich ascites mammary tumors and mitigated the proliferation of tumor cells in the peritoneal cavity in vivo. In summary, our findings demonstrate the high antitumor potential of PMB1.
Collapse
Affiliation(s)
- Deepa R. Bandi
- Department of Applied Microbiology, Sri Padmavathi Mahila Viswavidyalayam, Tirupati 517502, Andhra Pradesh, India; (D.R.B.); (P.S.D.)
| | - Ch M. Kumari Chitturi
- Department of Applied Microbiology, Sri Padmavathi Mahila Viswavidyalayam, Tirupati 517502, Andhra Pradesh, India; (D.R.B.); (P.S.D.)
| | - Jamuna Bai Aswathanarayan
- Department of Microbiology, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India;
| | - Prashant Kumar M. Veeresh
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India; (P.K.M.V.); (V.R.B.); (S.M.N.)
| | - Venugopal R. Bovilla
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India; (P.K.M.V.); (V.R.B.); (S.M.N.)
| | - Olga A. Sukocheva
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia;
| | - Potireddy Suvarnalatha Devi
- Department of Applied Microbiology, Sri Padmavathi Mahila Viswavidyalayam, Tirupati 517502, Andhra Pradesh, India; (D.R.B.); (P.S.D.)
| | - Suma M. Natraj
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India; (P.K.M.V.); (V.R.B.); (S.M.N.)
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India; (P.K.M.V.); (V.R.B.); (S.M.N.)
- Special Interest Group (SIG) in Cancer Biology and Cancer Stem Cells (CBCSC), JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India
| |
Collapse
|
16
|
Melnik BC, Stadler R, Weiskirchen R, Leitzmann C, Schmitz G. Potential Pathogenic Impact of Cow’s Milk Consumption and Bovine Milk-Derived Exosomal MicroRNAs in Diffuse Large B-Cell Lymphoma. Int J Mol Sci 2023; 24:ijms24076102. [PMID: 37047075 PMCID: PMC10094152 DOI: 10.3390/ijms24076102] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/05/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Epidemiological evidence supports an association between cow’s milk consumption and the risk of diffuse large B-cell lymphoma (DLBCL), the most common non-Hodgkin lymphoma worldwide. This narrative review intends to elucidate the potential impact of milk-related agents, predominantly milk-derived exosomes (MDEs) and their microRNAs (miRs) in lymphomagenesis. Upregulation of PI3K-AKT-mTORC1 signaling is a common feature of DLBCL. Increased expression of B cell lymphoma 6 (BCL6) and suppression of B lymphocyte-induced maturation protein 1 (BLIMP1)/PR domain-containing protein 1 (PRDM1) are crucial pathological deviations in DLBCL. Translational evidence indicates that during the breastfeeding period, human MDE miRs support B cell proliferation via epigenetic upregulation of BCL6 (via miR-148a-3p-mediated suppression of DNA methyltransferase 1 (DNMT1) and miR-155-5p/miR-29b-5p-mediated suppression of activation-induced cytidine deaminase (AICDA) and suppression of BLIMP1 (via MDE let-7-5p/miR-125b-5p-targeting of PRDM1). After weaning with the physiological termination of MDE miR signaling, the infant’s BCL6 expression and B cell proliferation declines, whereas BLIMP1-mediated B cell maturation for adequate own antibody production rises. Because human and bovine MDE miRs share identical nucleotide sequences, the consumption of pasteurized cow’s milk in adults with the continued transfer of bioactive bovine MDE miRs may de-differentiate B cells back to the neonatal “proliferation-dominated” B cell phenotype maintaining an increased BLC6/BLIMP1 ratio. Persistent milk-induced epigenetic dysregulation of BCL6 and BLIMP1 expression may thus represent a novel driving mechanism in B cell lymphomagenesis. Bovine MDEs and their miR cargo have to be considered potential pathogens that should be removed from the human food chain.
Collapse
|
17
|
Kato K, Fujii N, Makita S, Goto H, Kanda J, Shimada K, Akashi K, Izutsu K, Teshima T, Fukuda N, Sumitani T, Nakamura S, Sumi H, Shimizu S, Kakurai Y, Yoshikawa K, Tobinai K, Usui N, Hatake K. A phase 2 study of axicabtagene ciloleucel in relapsed or refractory large B-cell lymphoma in Japan: 1-year follow-up and biomarker analysis. Int J Hematol 2023; 117:409-420. [PMID: 36399286 DOI: 10.1007/s12185-022-03494-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
Abstract
Axicabtagene ciloleucel (axi-cel) is an autologous, CD19-targeting chimeric antigen receptor T‑cell therapy. We recently reported the 3-month follow-up results of a phase 2, multicenter, open‑label, single-arm study of axi-cel in Japanese patients with relapsed or refractory (R/R) large B-cell lymphoma (LBCL) (JapicCTI-183914). Here, we present 1-year efficacy and safety data and biomarker analysis data regarding mechanisms of resistance to axi-cel. Primary and secondary endpoints included investigator-assessed objective response rate (ORR), serious adverse events, and treatment-emergent adverse events. Axi-cel pharmacokinetics were also examined. Biomarker analysis was performed by cytokine measurement, immunohistochemistry, RNA sequencing, and whole-exome sequencing. At a median follow-up of 13.4 months, ORR was 86.7% (13/15 patients), and the complete response (CR) rate improved to 53.3% (8/15 patients) due to response conversion. Seven patients experienced disease progression, and one achieved CR after re-treatment with axi-cel. No new safety concerns were detected. Plausible resistance mechanisms to axi-cel varied among patients but included CD19 downregulation, programmed death-ligand 1 upregulation, and increased macrophage and angiogenesis signatures. The 1-year efficacy and safety of axi-cel were confirmed in Japanese patients with R/R LBCL. Resistance to treatment may involve multiple factors, including target antigen loss and an unfavorable tumor environment.Clinical trial registration: Japan Clinical Trials Information; JapicCTI-183914.
Collapse
Affiliation(s)
- Koji Kato
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Fukuoka Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Nobuharu Fujii
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Shinichi Makita
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
| | - Hideki Goto
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Junya Kanda
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuyuki Shimada
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Fukuoka Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Izutsu
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | - Kensei Tobinai
- Geriatric Health Services Facility Rehabilitation Care Funabashi, Chiba, Japan
| | - Noriko Usui
- Department of Clinical Oncology and Hematology, The Jikei University Daisan Hospital, Tokyo, Japan
| | - Kiyohiko Hatake
- Department of Lymphoma/Hematology Center, Mita Hospital, International University of Health and Welfare, Tokyo, Japan
| |
Collapse
|
18
|
Khan SU, Khan IM, Khan MU, Ud Din MA, Khan MZ, Khan NM, Liu Y. Role of LGMN in tumor development and its progression and connection with the tumor microenvironment. Front Mol Biosci 2023; 10:1121964. [PMID: 36825203 PMCID: PMC9942682 DOI: 10.3389/fmolb.2023.1121964] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
Legumain (LGMN) has been demonstrated to be overexpressed not just in breast, prostatic, and liver tumor cells, but also in the macrophages that compose the tumor microenvironment. This supports the idea that LGMN is a pivotal protein in regulating tumor development, invasion, and dissemination. Targeting LGMN with siRNA or chemotherapeutic medicines and peptides can suppress cancer cell proliferation in culture and reduce tumor growth in vivo. Furthermore, legumain can be used as a marker for cancer detection and targeting due to its expression being significantly lower in normal cells compared to tumors or tumor-associated macrophages (TAMs). Tumor formation is influenced by aberrant expression of proteins and alterations in cellular architecture, but the tumor microenvironment is a crucial deciding factor. Legumain (LGMN) is an in vivo-active cysteine protease that catalyzes the degradation of numerous proteins. Its precise biological mechanism encompasses a number of routes, including effects on tumor-associated macrophage and neovascular endothelium in the tumor microenvironment. The purpose of this work is to establish a rationale for thoroughly investigating the function of LGMN in the tumor microenvironment and discovering novel tumor early diagnosis markers and therapeutic targets by reviewing the function of LGMN in tumor genesis and progression and its relationship with tumor milieu.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China,Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China,*Correspondence: Ibrar Muhammad Khan, ; Yong Liu,
| | - Munir Ullah Khan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, International Research Center for X Polymers, Zhejiang University, Hangzhou, China
| | - Muhammad Azhar Ud Din
- Faculty of Pharmacy, Gomal University Dera Ismail Khan KPK, Dera IsmailKhan, Pakistan
| | - Muhammad Zahoor Khan
- Department of Animal Breeding and Genetics, Faculty of Veterinary and Animal Sciences, University of Agriculture, Dera IsmailKhan, Pakistan
| | - Nazir Muhammad Khan
- Department of Zoology, University of Science and Technology, Bannu, Pakistan
| | - Yong Liu
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China,*Correspondence: Ibrar Muhammad Khan, ; Yong Liu,
| |
Collapse
|
19
|
Ng WL, Ansell SM, Mondello P. Insights into the tumor microenvironment of B cell lymphoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:362. [PMID: 36578079 PMCID: PMC9798587 DOI: 10.1186/s13046-022-02579-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/20/2022] [Indexed: 12/30/2022]
Abstract
The standard therapies in lymphoma have predominantly focused on targeting tumor cells with less of a focus on the tumor microenvironment (TME), which plays a critical role in favoring tumor growth and survival. Such an approach may result in increasingly refractory disease with progressively reduced responses to subsequent treatments. To overcome this hurdle, targeting the TME has emerged as a new therapeutic strategy. The TME consists of T and B lymphocytes, tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), and other components. Understanding the TME can lead to a comprehensive approach to managing lymphoma, resulting in therapeutic strategies that target not only cancer cells, but also the supportive environment and thereby ultimately improve survival of lymphoma patients. Here, we review the normal function of different components of the TME, the impact of their aberrant behavior in B cell lymphoma and the current TME-direct therapeutic avenues.
Collapse
Affiliation(s)
- Wern Lynn Ng
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Stephen M. Ansell
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Patrizia Mondello
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| |
Collapse
|
20
|
Lin SC, Liao YC, Chen PM, Yang YY, Wang YH, Tung SL, Chuang CM, Sung YW, Jang TH, Chuang SE, Wang LH. Periostin promotes ovarian cancer metastasis by enhancing M2 macrophages and cancer-associated fibroblasts via integrin-mediated NF-κB and TGF-β2 signaling. J Biomed Sci 2022; 29:109. [PMID: 36550569 PMCID: PMC9784270 DOI: 10.1186/s12929-022-00888-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Ovarian cancer has the highest mortality among gynecological cancers due to late diagnosis and lack of effective targeted therapy. Although the study of interplay between cancer cells with their microenvironment is emerging, how ovarian cancer triggers signaling that coordinates with immune cells to promote metastasis is still elusive. METHODS Microarray and bioinformatics analysis of low and highly invasive ovarian cancer cell lines were used to reveal periostin (POSTN), a matrix protein with multifunctions in cancer, with elevated expression in the highly invasive cells. Anchorage independent assay, Western blot, RNA interference, confocal analysis and neutralizing antibody treatment were performed to analyze the effects of POSTN on tumor promotion and to explore the underlying mechanism. Chemotaxis, flow cytometry and cytokine array analyses were undertaken to analyze the involvement of POSTN in cancer-associated fibroblast (CAF) and macrophage modulation. Correlations between POSTN expression levels and clinical characteristics were analyzed using the Oncomine, commercial ovarian cancer cDNA and China Medical University Hospital patient cohort. In vivo effect of POSTN on metastasis was studied using a mouse xenograft model. RESULTS Expression of POSTN was found to be elevated in highly invasive ovarian cancer cells. We observed that POSTN was co-localized with integrin β3 and integrin β5, which was important for POSTN-mediated activation of ERK and NF-κB. Ectopic expression of POSTN enhanced whereas knockdown of POSTN decreased cancer cell migration and invasion in vitro, as well as tumor growth and metastasis in vivo. POSTN enhanced integrin/ERK/NF-κB signaling through an autocrine effect on cancer cells to produce macrophage attracting and mobilizing cytokines including MIP-1β, MCP-1, TNFα and RANTES resulting in increased chemotaxis of THP-1 monocytes and their polarization to M2 macrophages in vitro. In agreement, tumors derived from POSTN-overexpressing SKOV3 harbored more tumor-associated macrophages than the control tumors. POSTN induced TGF-β2 expression from ovarian cancer cells to promote activation of adipose-derived stromal cells to become CAF-like cells expressing alpha smooth muscle actin and fibroblast activation protein alpha. Consistently, increased CAFs were observed in POSTN overexpressing SKOV3 cells-derived metastatic tumors. In clinical relevance, we found that expression of POSTN was positively correlated with advanced-stage diseases and poor overall survival of patients. CONCLUSIONS Our study revealed a POSTN-integrin-NF-κB-mediated signaling and its involvement in enhancing M2 macrophages and CAFs, which could potentially participate in promoting tumor growth. Our results suggest that POSTN could be a useful prognosis marker and potential therapeutic target.
Collapse
Affiliation(s)
- Sheng-Chieh Lin
- grid.254145.30000 0001 0083 6092Graduate Institute of Integrated Medicine and Chinese Medicine Research Center, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402 Taiwan ,grid.254145.30000 0001 0083 6092Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Yi-Chu Liao
- grid.59784.370000000406229172Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Po-Ming Chen
- grid.254145.30000 0001 0083 6092Graduate Institute of Integrated Medicine and Chinese Medicine Research Center, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402 Taiwan ,grid.452796.b0000 0004 0634 3637Research Assistant Center, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Ya-Yu Yang
- grid.59784.370000000406229172National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yi-Hsiang Wang
- grid.59784.370000000406229172Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan ,grid.38348.340000 0004 0532 0580Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Shiao-Lin Tung
- Department of Hematology and Oncology, Ton-Yen General Hospital, Hsinchu, Taiwan ,Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan, Taiwan
| | - Chi-Mu Chuang
- grid.278247.c0000 0004 0604 5314Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan ,grid.260539.b0000 0001 2059 7017School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Wen Sung
- grid.254145.30000 0001 0083 6092Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan ,grid.411508.90000 0004 0572 9415Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Te-Hsuan Jang
- grid.59784.370000000406229172National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan ,grid.38348.340000 0004 0532 0580Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Shuang-En Chuang
- grid.59784.370000000406229172National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Lu-Hai Wang
- grid.254145.30000 0001 0083 6092Graduate Institute of Integrated Medicine and Chinese Medicine Research Center, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402 Taiwan ,grid.59784.370000000406229172Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan ,grid.38348.340000 0004 0532 0580Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
21
|
Solberg R, Lunde NN, Forbord KM, Okla M, Kassem M, Jafari A. The Mammalian Cysteine Protease Legumain in Health and Disease. Int J Mol Sci 2022; 23:ijms232415983. [PMID: 36555634 PMCID: PMC9788469 DOI: 10.3390/ijms232415983] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
The cysteine protease legumain (also known as asparaginyl endopeptidase or δ-secretase) is the only known mammalian asparaginyl endopeptidase and is primarily localized to the endolysosomal system, although it is also found extracellularly as a secreted protein. Legumain is involved in the regulation of diverse biological processes and tissue homeostasis, and in the pathogenesis of various malignant and nonmalignant diseases. In addition to its proteolytic activity that leads to the degradation or activation of different substrates, legumain has also been shown to have a nonproteolytic ligase function. This review summarizes the current knowledge about legumain functions in health and disease, including kidney homeostasis, hematopoietic homeostasis, bone remodeling, cardiovascular and cerebrovascular diseases, fibrosis, aging and senescence, neurodegenerative diseases and cancer. In addition, this review addresses the effects of some marketed drugs on legumain. Expanding our knowledge on legumain will delineate the importance of this enzyme in regulating physiological processes and disease conditions.
Collapse
Affiliation(s)
- Rigmor Solberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
- Correspondence: (R.S.); (A.J.); Tel.: +47-22-857-514 (R.S.); +45-35-337-423 (A.J.)
| | - Ngoc Nguyen Lunde
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
| | - Karl Martin Forbord
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
| | - Meshail Okla
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Abbas Jafari
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Correspondence: (R.S.); (A.J.); Tel.: +47-22-857-514 (R.S.); +45-35-337-423 (A.J.)
| |
Collapse
|
22
|
Fortis SP, Sofopoulos M, Goulielmaki M, Arnogiannaki N, Ardavanis A, Perez SA, Gritzapis AD, Baxevanis CN. Association between Intratumoral CD8+ T Cells with FoxP3+ and CD163+ Cells: A Potential Immune Intrinsic Negative Feedback Mechanism for Acquired Immune Resistance. Cancers (Basel) 2022; 14:cancers14246208. [PMID: 36551693 PMCID: PMC9777444 DOI: 10.3390/cancers14246208] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Acquired immune resistance (AIR) describes a situation in which cancer patients who initially responded clinically to immunotherapies, after a certain period of time, progress with their disease. Considering that AIR represents a feedback response of the tumor against the immune attack generated during the course of immunotherapies, it is conceivable that AIR may also occur before treatment initiation as a mechanism to escape endogenous adaptive antitumor immunity (EAAI). In the present study, we assessed the EAAI in paraffin-embedded breast primary tumor tissue samples and drew correlations with the clinical outcomes. In particular, we analyzed densities of CD8+ cells as elements mediating antitumor cytotoxicity, and of CD163+ and FoxP3+ cells as suppressor elements. We found a direct correlation between the densities of CD8+ cells and of CD163+ and/or FoxP3+ cells in the vast majority of patients' tumors. Importantly, the vast majority of patients whose tumors were overpopulated by CD8+ cells developed AIR, which was characterized by high intratumoral CD163+ and/or FoxP3+ cell densities and reduced overall survival (OS). We also showed that AIR depends on the levels of CD8+ cell-ratios in the tumor center to the invasive margin. Our data suggest that tumors develop AIR only when under a robust endogenous immune pressure.
Collapse
Affiliation(s)
- Sotirios P. Fortis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | | | - Maria Goulielmaki
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Niki Arnogiannaki
- Pathology Department, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Alexandros Ardavanis
- First Medical Oncology Clinic, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Sonia A. Perez
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Angelos D. Gritzapis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Constantin N. Baxevanis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
- Correspondence: ; Tel.: +30-21064-09380
| |
Collapse
|
23
|
Zeller T, Lutz S, Münnich IA, Windisch R, Hilger P, Herold T, Tahiri N, Banck JC, Weigert O, Moosmann A, von Bergwelt-Baildon M, Flamann C, Bruns H, Wichmann C, Baumann N, Valerius T, Schewe DM, Peipp M, Rösner T, Humpe A, Kellner C. Dual checkpoint blockade of CD47 and LILRB1 enhances CD20 antibody-dependent phagocytosis of lymphoma cells by macrophages. Front Immunol 2022; 13:929339. [PMID: 36389667 PMCID: PMC9647079 DOI: 10.3389/fimmu.2022.929339] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/12/2022] [Indexed: 11/28/2022] Open
Abstract
Antibody-dependent cellular phagocytosis (ADCP) by macrophages, an important effector function of tumor targeting antibodies, is hampered by ‘Don´t Eat Me!’ signals such as CD47 expressed by cancer cells. Yet, human leukocyte antigen (HLA) class I expression may also impair ADCP by engaging leukocyte immunoglobulin-like receptor subfamily B (LILRB) member 1 (LILRB1) or LILRB2. Analysis of different lymphoma cell lines revealed that the ratio of CD20 to HLA class I cell surface molecules determined the sensitivity to ADCP by the combination of rituximab and an Fc-silent variant of the CD47 antibody magrolimab (CD47-IgGσ). To boost ADCP, Fc-silent antibodies against LILRB1 and LILRB2 were generated (LILRB1-IgGσ and LILRB2-IgGσ, respectively). While LILRB2-IgGσ was not effective, LILRB1-IgGσ significantly enhanced ADCP of lymphoma cell lines when combined with both rituximab and CD47-IgGσ. LILRB1-IgGσ promoted serial engulfment of lymphoma cells and potentiated ADCP by non-polarized M0 as well as polarized M1 and M2 macrophages, but required CD47 co-blockade and the presence of the CD20 antibody. Importantly, complementing rituximab and CD47-IgGσ, LILRB1-IgGσ increased ADCP of chronic lymphocytic leukemia (CLL) or lymphoma cells isolated from patients. Thus, dual checkpoint blockade of CD47 and LILRB1 may be promising to improve antibody therapy of CLL and lymphomas through enhancing ADCP by macrophages.
Collapse
Affiliation(s)
- Tobias Zeller
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Lutz
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Ira A. Münnich
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Roland Windisch
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Patricia Hilger
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Tobias Herold
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Natyra Tahiri
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Jan C. Banck
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Oliver Weigert
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Moosmann
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- DZIF – German Center for Infection Research, Munich, Germany
- Helmholtz Zentrum München, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cindy Flamann
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Heiko Bruns
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Christian Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Niklas Baumann
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Denis M. Schewe
- Department of Pediatrics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thies Rösner
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Andreas Humpe
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Christian Kellner
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
- *Correspondence: Christian Kellner,
| |
Collapse
|
24
|
Xie Y, Yang H, Yang C, He L, Zhang X, Peng L, Zhu H, Gao L. Role and Mechanisms of Tumor-Associated Macrophages in Hematological Malignancies. Front Oncol 2022; 12:933666. [PMID: 35875135 PMCID: PMC9301190 DOI: 10.3389/fonc.2022.933666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Mounting evidence has revealed that many nontumor cells in the tumor microenvironment, such as fibroblasts, endothelial cells, mesenchymal stem cells, and leukocytes, are strongly involved in tumor progression. In hematological malignancies, tumor-associated macrophages (TAMs) are considered to be an important component that promotes tumor growth and can be polarized into different phenotypes with protumor or antitumor roles. This Review emphasizes research related to the role and mechanisms of TAMs in hematological malignancies. TAMs lead to poor prognosis by influencing tumor progression at the molecular level, including nurturing cancer stem cells and laying the foundation for metastasis. Although detailed molecular mechanisms have not been clarified, TAMs may be a new therapeutic target in hematological disease treatment.
Collapse
|
25
|
Zhang M, Zhu J, Wang W, Jiang Z. Active legumain promotes invasion and migration of neuroblastoma by regulating epithelial-mesenchymal transition. Open Life Sci 2022; 17:676-685. [PMID: 35800070 PMCID: PMC9214917 DOI: 10.1515/biol-2022-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 11/26/2021] [Accepted: 01/03/2022] [Indexed: 11/18/2022] Open
Abstract
Neuroblastoma (NB) is a commonly occurring malignancy in children. Epithelial-mesenchymal transition (EMT) is an adaptive change in promoting tumor metastasis. As an important factor in regulating tumor metastasis, whether legumain could promote metastasis of NB by EMT is still unexplored. Legumain is the active form of prolegumain, abundant in tumor plasma. So in the current study, different forms of legumain were identified in NB. Second, correlation analysis of N-cadherin and active legumain was identified by western blot analysis. Third, legumain gene amplification or gene knockdown were proceeded to examine the effect of legumain on EMT by scratch and transwell assay; meanwhile, active mature legumain or its asparagine endopeptidase (AEP) inhibitor was also added in. Finally, legumain can be detected differently in NB cells. Changes in legumain could influence NB metastasis by regulating EMT markers (e.g., N-cadherin, vimentin, and slug). Besides, the effect of legumain on EMT by its AEP activity was proved by intervention experiment of AEP gene transfection and gene knockdown experiments or adding recombinant human legumain suspension or specific inhibitor of AEP in NB cells (p < 0.05). These results suggest that legumain can promote invasion and migration of NB by regulating EMT, and EMT of NB is regulated by AEP activity of legumain, which can be inhibited by a specific AEP inhibitor.
Collapse
Affiliation(s)
- Min Zhang
- Department of Emergency & Trauma Surgery, Shanghai University of Medicine and Health Sciences, Affiliated Zhoupu Hospital , Shanghai 201318 , P. R. China
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200092 , P. R. China
| | - Jianhua Zhu
- Department of Emergency & Trauma Surgery, Shanghai University of Medicine and Health Sciences, Affiliated Zhoupu Hospital , Shanghai 201318 , P. R. China
| | - Wei Wang
- Department of Emergency & Trauma Surgery, Shanghai University of Medicine and Health Sciences, Affiliated Zhoupu Hospital , Shanghai 201318 , P. R. China
| | - Zhiteng Jiang
- Colloge of Pharmacy, Shanghai University of Medicine and Health Sciences , Shanghai 201318 , P. R. China
| |
Collapse
|
26
|
Asparaginyl endopeptidase protects against podocyte injury in diabetic nephropathy through cleaving cofilin-1. Cell Death Dis 2022; 13:184. [PMID: 35217650 PMCID: PMC8881581 DOI: 10.1038/s41419-022-04621-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/31/2022] [Accepted: 02/08/2022] [Indexed: 11/08/2022]
Abstract
AbstractPodocyte injury and loss are critical events in diabetic nephropathy (DN); however, the underlying molecular mechanisms remain unclear. Here, we demonstrate that asparaginyl endopeptidase (AEP) protects against podocyte injury through modulating the dynamics of the cytoskeleton. AEP was highly upregulated in diabetic glomeruli and hyperglycemic stimuli treated-podocytes; however, AEP gene knockout and its compound inhibitor treatment accelerated DN in streptozotocin-induced diabetic mice, whereas specific induction of AEP in glomerular cells attenuated podocyte injury and renal function deterioration. In vitro, elevated AEP was involved in actin cytoskeleton maintenance and anti-apoptosis effects. Mechanistically, we found that AEP directly cleaved the actin-binding protein cofilin-1 after the asparagine 138 (N138) site. The protein levels of endogenous cofilin-1 1-138 fragments were upregulated in diabetic podocytes, consistent with the changes in AEP levels. Importantly, we found that cofilin-1 1-138 fragments were remarkably unphosphorylated than full-length cofilin-1, indicating the enhanced cytoskeleton maintenance activity of cofilin-1 1-138. Then we validated cofilin-1 1-138 could rescue podocytes from cytoskeleton disarrangement and injury in diabetic conditions. Taken together, our data suggest a protective role of elevated AEP in podocyte injury during DN progression through cleaving cofilin-1 to maintain podocyte cytoskeleton dynamics and defend damage.
Collapse
|
27
|
Serganova I, Chakraborty S, Yamshon S, Isshiki Y, Bucktrout R, Melnick A, Béguelin W, Zappasodi R. Epigenetic, Metabolic, and Immune Crosstalk in Germinal-Center-Derived B-Cell Lymphomas: Unveiling New Vulnerabilities for Rational Combination Therapies. Front Cell Dev Biol 2022; 9:805195. [PMID: 35071240 PMCID: PMC8777078 DOI: 10.3389/fcell.2021.805195] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/30/2021] [Indexed: 12/24/2022] Open
Abstract
B-cell non-Hodgkin lymphomas (B-NHLs) are highly heterogenous by genetic, phenotypic, and clinical appearance. Next-generation sequencing technologies and multi-dimensional data analyses have further refined the way these diseases can be more precisely classified by specific genomic, epigenomic, and transcriptomic characteristics. The molecular and genetic heterogeneity of B-NHLs may contribute to the poor outcome of some of these diseases, suggesting that more personalized precision-medicine approaches are needed for improved therapeutic efficacy. The germinal center (GC) B-cell like diffuse large B-cell lymphomas (GCB-DLBCLs) and follicular lymphomas (FLs) share specific epigenetic programs. These diseases often remain difficult to treat and surprisingly do not respond advanced immunotherapies, despite arising in secondary lymphoid organs at sites of antigen recognition. Epigenetic dysregulation is a hallmark of GCB-DLBCLs and FLs, with gain-of-function (GOF) mutations in the histone methyltransferase EZH2, loss-of-function (LOF) mutations in histone acetyl transferases CREBBP and EP300, and the histone methyltransferase KMT2D representing the most prevalent genetic lesions driving these diseases. These mutations have the common effect to disrupt the interactions between lymphoma cells and the immune microenvironment, via decreased antigen presentation and responsiveness to IFN-γ and CD40 signaling pathways. This indicates that immune evasion is a key step in GC B-cell lymphomagenesis. EZH2 inhibitors are now approved for the treatment of FL and selective HDAC3 inhibitors counteracting the effects of CREBBP LOF mutations are under development. These treatments can help restore the immune control of GCB lymphomas, and may represent optimal candidate agents for more effective combination with immunotherapies. Here, we review recent progress in understanding the impact of mutant chromatin modifiers on immune evasion in GCB lymphomas. We provide new insights on how the epigenetic program of these diseases may be regulated at the level of metabolism, discussing the role of metabolic intermediates as cofactors of epigenetic enzymes. In addition, lymphoma metabolic adaptation can negatively influence the immune microenvironment, further contributing to the development of immune cold tumors, poorly infiltrated by effector immune cells. Based on these findings, we discuss relevant candidate epigenetic/metabolic/immune targets for rational combination therapies to investigate as more effective precision-medicine approaches for GCB lymphomas.
Collapse
Affiliation(s)
- Inna Serganova
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Sanjukta Chakraborty
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Samuel Yamshon
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Yusuke Isshiki
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Ryan Bucktrout
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Ari Melnick
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Wendy Béguelin
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Roberta Zappasodi
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States.,Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| |
Collapse
|
28
|
Comparetti EJ, Ferreira NN, Ferreira LMB, Kaneno R, Zucolotto V. Immunomodulatory properties of nanostructured systems for cancer therapy. J Biomed Mater Res A 2022; 110:1166-1181. [PMID: 35043549 DOI: 10.1002/jbm.a.37359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/06/2021] [Accepted: 01/03/2022] [Indexed: 11/10/2022]
Abstract
Based on statistical data reported in 2020, cancer was responsible for approximately 10 million deaths. Furthermore, 17 million new cases were diagnosed worldwide. Nanomedicine and immunotherapy have shown satisfactory clinical results among all scientific and technological alternatives for the treatment of cancer patients. Immunotherapy-based treatments comprise the consideration of new alternatives to hinder neoplastic proliferation and to reduce adverse events in the body, thereby promoting immune destruction of diseased cells. Additionally, nanostructured systems have been proven to elicit specific immune responses that may enhance anti-tumor activity. A new generation of nanomedicines, based on biomimetic and bioinspired systems, has been proposed to target tumors by providing immunomodulatory features and by enabling recovery of human immune destruction capacity against cancer cells. This review provides an overview of the aspects and the mechanisms by which nanomedicines can be used to enhance clinical procedures using the immune modulatory responses of nanoparticles (NPs) in the host defense system. We initially outline the cancer statistics for conventional and new treatment approaches providing a brief description of the human host defense system and basic principles of NP interactions with monocytes, leukocytes, and dendritic cells for the modulation of antitumor immune responses. A report on different biomimetic and bioinspired systems is also presented here and their particularities in cancer treatments are addressed, highlighting their immunomodulatory properties. Finally, we propose future perspectives regarding this new therapeutic strategy, highlighting the main challenges for future use in clinical practice.
Collapse
Affiliation(s)
- Edson J Comparetti
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Natalia N Ferreira
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Leonardo M B Ferreira
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Ramon Kaneno
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, Brazil
| | - Valtencir Zucolotto
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, Brazil
| |
Collapse
|
29
|
Rømer AMA, Thorseth ML, Madsen DH. Immune Modulatory Properties of Collagen in Cancer. Front Immunol 2021; 12:791453. [PMID: 34956223 PMCID: PMC8692250 DOI: 10.3389/fimmu.2021.791453] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
During tumor growth the extracellular matrix (ECM) undergoes dramatic remodeling. The normal ECM is degraded and substituted with a tumor-specific ECM, which is often of higher collagen density and increased stiffness. The structure and collagen density of the tumor-specific ECM has been associated with poor prognosis in several types of cancer. However, the reason for this association is still largely unknown. Collagen can promote cancer cell growth and migration, but recent studies have shown that collagens can also affect the function and phenotype of various types of tumor-infiltrating immune cells such as tumor-associated macrophages (TAMs) and T cells. This suggests that tumor-associated collagen could have important immune modulatory functions within the tumor microenvironment, affecting cancer progression as well as the efficacy of cancer immunotherapy. The effects of tumor-associated collagen on immune cells could help explain why a high collagen density in tumors is often correlated with a poor prognosis. Knowledge about immune modulatory functions of collagen could potentially identify targets for improving current cancer therapies or for development of new treatments. In this review, the current knowledge about the ability of collagen to influence T cell activity will be summarized. This includes direct interactions with T cells as well as induction of immune suppressive activity in other immune cells such as macrophages. Additionally, the potential effects of collagen on the efficacy of cancer immunotherapy will be discussed.
Collapse
Affiliation(s)
- Anne Mette Askehøj Rømer
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Marie-Louise Thorseth
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Hargbøl Madsen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Welte T, Mai J, Zhang Z, Tian S, Zhang G, Xu Y, Zhang L, Chen SS, Wang T, Shen H. A heparan-sulfate-bearing syndecan-1 glycoform is a distinct surface marker for intra-tumoral myeloid-derived suppressor cells. iScience 2021; 24:103349. [PMID: 34825135 PMCID: PMC8603209 DOI: 10.1016/j.isci.2021.103349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/17/2021] [Accepted: 10/22/2021] [Indexed: 11/19/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) infiltrate cancer tissue, promote tumor growth, and are associated with resistance to cancer therapies. However, there is no practical approach available to distinguish MDSCs from mature counterparts inside tumors. Here, we show that a recently isolated thioaptamer probe (T1) binds to MDSC subsets in colorectal and pancreatic tumors with high specificity. Whole transcriptome and functional analysis revealed that T1-binding cells contain polymorphonuclear (PMN)-MDSCs characterized by several immunosuppression pathways, ROS production, and T cell suppression activity, whereas T1-non-binding PMNs were mature and nonsuppressive. We identified syndecan-1 as the T1-interacting protein on MDSCs and chronic myelogenous leukemia K562 cell line. Heparan sulfate chains were essential in T1-binding. Inside tumors PMN-MDSCs expressed heparan sulfate biogenesis enzymes at higher levels. Tumor-cell-derived soluble factor(s) enhanced MDSCs' affinity for T1. Overall, we uncovered heparan-sulfate-dependent MDSC modulation in the tumor microenvironment and identified T1 as tool preferentially targeting tumor-promoting myeloid cell subsets.
Collapse
Affiliation(s)
- Thomas Welte
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Zhe Zhang
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Shaohui Tian
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Guodong Zhang
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Yitian Xu
- Center for Immunotherapy Research, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Licheng Zhang
- Center for Immunotherapy Research, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Shu-shia Chen
- Center for Immunotherapy Research, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Tian Wang
- Department of Microbiology & Immunology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
- Innovative Therapeutic Program, Houston Methodist Cancer Center, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
31
|
Recapitulating the Angiogenic Switch in a Hydrogel-Based 3D In Vitro Tumor-Stroma Model. Bioengineering (Basel) 2021; 8:bioengineering8110186. [PMID: 34821752 PMCID: PMC8614676 DOI: 10.3390/bioengineering8110186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/02/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
To ensure nutrient and oxygen supply, tumors beyond a size of 1–2 mm3 need a connection to the vascular system. Thus, tumor cells modify physiological tissue homeostasis by secreting inflammatory and angiogenic cytokines. This leads to the activation of the tumor microenvironment and the turning of the angiogenic switch, resulting in tumor vascularization and growth. To inhibit tumor growth by developing efficient anti-angiogenic therapies, an in depth understanding of the molecular mechanism initiating angiogenesis is essential. Yet so far, predominantly 2D cell cultures or animal models have been used to clarify the interactions within the tumor stroma, resulting in poor transferability of the data obtained to the in vivo situation. Consequently, there is an abundant need for complex, humanized, 3D models in vitro. We established a dextran-hydrogel-based 3D organotypic in vitro model containing microtumor spheroids, macrophages, neutrophils, fibroblasts and endothelial cells, allowing for the analysis of tumor–stroma interactions in a controlled and modifiable environment. During the cultivation period of 21 days, the microtumor spheroids in the model grew in size and endothelial cells formed elongated tubular structures resembling capillary vessels, that appeared to extend towards the tumor spheroids. The tubular structures exhibited complex bifurcations and expanded without adding external angiogenic factors such as VEGF to the culture. To allow high-throughput screening of therapeutic candidates, the 3D cell culture model was successfully miniaturized to a 96-well format, while still maintaining the same level of tumor spheroid growth and vascular sprouting. The quantification of VEGF in the conditioned medium of these cultures showed a continuous increase during the cultivation period, suggesting the contribution of endogenous VEGF to the induction of the angiogenic switch and vascular sprouting. Thus, this model is highly suitable as a testing platform for novel anticancer therapeutics targeting the tumor as well as the vascular compartment.
Collapse
|
32
|
Liu Y, Zhou X, Wang X. Targeting the tumor microenvironment in B-cell lymphoma: challenges and opportunities. J Hematol Oncol 2021; 14:125. [PMID: 34404434 PMCID: PMC8369706 DOI: 10.1186/s13045-021-01134-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
B-cell lymphoma is a group of hematological malignancies with high clinical and biological heterogeneity. The pathogenesis of B-cell lymphoma involves a complex interaction between tumor cells and the tumor microenvironment (TME), which is composed of stromal cells and extracellular matrix. Although the roles of the TME have not been fully elucidated, accumulating evidence implies that TME is closely relevant to the origination, invasion and metastasis of B-cell lymphoma. Explorations of the TME provide distinctive insights for cancer therapy. Here, we epitomize the recent advances of TME in B-cell lymphoma and discuss its function in tumor progression and immune escape. In addition, the potential clinical value of targeting TME in B-cell lymphoma is highlighted, which is expected to pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Yingyue Liu
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324, Jingwu Road, Jinan, 250021, Shandong, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- School of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- School of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
33
|
Vázquez S, Vallejo R, Espinosa J, Arteche N, Vega JA, Pérez V. Immunohistochemical Characterization of Tumor-Associated Macrophages in Canine Lymphomas. Animals (Basel) 2021; 11:2301. [PMID: 34438760 PMCID: PMC8388421 DOI: 10.3390/ani11082301] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/23/2021] [Accepted: 08/01/2021] [Indexed: 11/17/2022] Open
Abstract
Macrophages have been confirmed to play a significant role in the behavior of human lymphomas, albeit no consistent data are so far available in canine lymphomas. The present study characterizes the macrophages present in cases of canine nodal lymphoma and their relationship with the histological grade and the immunophenotype. Samples from the lymph nodes of 25 dogs diagnosed with lymphoma were selected. Immunohistochemistry was used to determine the tumor immunophenotype (CD3 and CD20 antibodies) and macrophage characterization (Iba1, MAC387, CD204, CD163 and iNOS antibodies). Macrophage counting was performed in 10 randomly selected, high-power fields per sample. Generalized linear models with Poisson distribution were used for statistical analysis. A significantly greater number of macrophages (Iba1+) were detected in high-grade and B-cell lymphomas. The highest amount of both M1 (iNOS+) and M2 (CD204+ and CD163+) subtypes were observed in B-cell lymphomas. High-grade lymphomas showed a greater number of CD204+ and CD163+ cells and recently recruited MAC387+ macrophages. The latter were most abundant in T than in B-cell lymphomas. In conclusion, a significant population of macrophages is present in canine lymphomas, which constitute a heterogeneous population that shows variations in the amount and immunohistochemical profile according to the histological grade and immunophenotype.
Collapse
Affiliation(s)
- Sergio Vázquez
- Departamento de Sanidad Animal, Universidad de León, 24007 León, Spain; (R.V.); (J.E.); (N.A.); (V.P.)
| | - Raquel Vallejo
- Departamento de Sanidad Animal, Universidad de León, 24007 León, Spain; (R.V.); (J.E.); (N.A.); (V.P.)
| | - José Espinosa
- Departamento de Sanidad Animal, Universidad de León, 24007 León, Spain; (R.V.); (J.E.); (N.A.); (V.P.)
| | - Noive Arteche
- Departamento de Sanidad Animal, Universidad de León, 24007 León, Spain; (R.V.); (J.E.); (N.A.); (V.P.)
| | - José A. Vega
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain;
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Providencia 7500912, Santiago de Chile, Chile
| | - Valentín Pérez
- Departamento de Sanidad Animal, Universidad de León, 24007 León, Spain; (R.V.); (J.E.); (N.A.); (V.P.)
| |
Collapse
|
34
|
Cencini E, Fabbri A, Sicuranza A, Gozzetti A, Bocchia M. The Role of Tumor-Associated Macrophages in Hematologic Malignancies. Cancers (Basel) 2021; 13:cancers13143597. [PMID: 34298810 PMCID: PMC8304632 DOI: 10.3390/cancers13143597] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/11/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Tumor-associated macrophages (TAM) represent a leading component of the tumor microenvironment in hematologic malignancies. TAM could display antitumor activity or, conversely, could contribute to tumor growth and survival, depending on their polarization. TAM are polarized towards form M1, with a pro-inflammatory phenotype and an antineoplastic activity, or M2, with an alternately activated phenotype, associated with a poor outcome in patients presenting with leukemia, lymphoma or multiple myeloma. The molecular mechanisms of TAM in different types of hematologic malignancies are different due to the peculiar microenvironment of each disease. TAM could contribute to tumor progression, reduced apoptosis and angiogenesis; a different TAM polarization could explain a reduced treatment response in patients with a similar disease subtype. The aim of our review is to better define the role of TAM in patients with leukemia, lymphoma or multiple myeloma. Finally, we would like to focus on TAM as a possible target for antineoplastic therapy. Abstract The tumor microenvironment includes dendritic cells, T-cytotoxic, T-helper, reactive B-lymphoid cells and macrophages; these reactive cells could interplay with malignant cells and promote tumor growth and survival. Among its cellular components, tumor-associated macrophages (TAM) represent a component of the innate immune system and play an important role, especially in hematologic malignancies. Depending on the stimuli that trigger their activation, TAM are polarized towards form M1, contributing to antitumor responses, or M2, associated with tumor progression. Many studies demonstrated a correlation between TAM, disease progression and the patient’s outcome in lymphoproliferative neoplasms, such as Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL), even if with conflicting results. A critical hurdle to overcome is surely represented by the heterogeneity in the choice of the optimal markers and methods used for TAM analysis (gene-expression profile vs. immunohistochemistry, CD163vs. CD68vs. CD163/CD68 double-positive cells). TAM have been recently linked to the development and progression of multiple myeloma and leukemia, with a critical role in the homing of malignant cells, drug resistance, immune suppression and angiogenesis. As such, this review will summarize the role of TAM in different hematologic malignancies, focusing on the complex interplay between TAM and tumor cells, the prognostic value of TAM and the possible TAM-targeted therapeutic strategies.
Collapse
|
35
|
Tian WJ, Wang QN, Wang XF, Dong DF. Clophosome alleviate dextran sulphate sodium-induced colitis by regulating gut immune responses and maintaining intestinal integrity in mice. Clin Exp Pharmacol Physiol 2021; 48:902-910. [PMID: 33527445 DOI: 10.1111/1440-1681.13468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/12/2021] [Indexed: 01/08/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic progressive disorder characterized by complicated gastrointestinal inflammation. Research on therapeutic agents is still urgent due to the lack of satisfactory treatments. Gut macrophages are considered to be predominant in excessive inflammatory responses. Thus, we aimed to investigate whether depletion of macrophages would have a beneficial effect on IBD and could be a potential therapeutic strategy. In this study, we established a 12-day Dextran sodium sulphate (DSS)-induced colitis mouse model and determined the effect of the macrophage depletion agent Clophosome (neutral clodronate liposomes; CNC). The results showed that CNC significantly alleviated the symptoms of colitis, as demonstrated by greater weight gain, decreased disease activity index (DAI) scores, and lower histopathological damage scores, as well was reduced levels of the proinflammatory cytokines interleukin (IL)-6 and tumour necrosis factor (TNF)-α. To investigate T cell subsets, cells were isolated from the lamina propria and cultured to analyse the expression of IL-17A, interferon (IFN)-γ and Foxp3 in CD4+ cells by flow cytometry. The data showed that during the process of colitis, the frequencies of CD4+ IL-17A+ T cells were significantly increased. Notably, CNC treatment markedly reduced the population of CD4+ IL-17A+ T cells, especially CD4+ IL-17A+ IFN-γ+ T cells. Furthermore, intestinal barrier integrity, as assessed by immunostaining of mucin and tight junction proteins, was severely disrupted in colitis. CNC improved the intestinal barrier by enhancing the expression of muc-2 and occludin. In summary, our findings demonstrated that CNC successfully ameliorated DSS-induced colitis and that its effect may be associated with inhibiting inflammatory responses and maintaining intestinal integrity.
Collapse
Affiliation(s)
- Wen-Jie Tian
- Faculty of Medical Laboratory Science, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qian-Nan Wang
- Department of Pathology and Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xue-Feng Wang
- Faculty of Medical Laboratory Science, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dan-Feng Dong
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Integrative Statistics, Machine Learning and Artificial Intelligence Neural Network Analysis Correlated CSF1R with the Prognosis of Diffuse Large B-Cell Lymphoma. HEMATO 2021. [DOI: 10.3390/hemato2020011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor-associated macrophages (TAMs) of the immune microenvironment play an important role in the Diffuse Large B-cell Lymphoma (DLBCL) pathogenesis. This research aimed to characterize the expression of macrophage colony-stimulating factor 1 receptor (CSF1R) at the gene and protein level in correlation with survival. First, the immunohistochemical expression of CSF1R was analyzed in a series of 198 cases from Tokai University Hospital and two patterns of histological expression were found, a TAMs, and a diffuse B-lymphocytes pattern. The clinicopathological correlations showed that the CSF1R + TAMs pattern associated with a poor progression-free survival of the patients, disease progression, higher MYC proto-oncogene expression, lower MDM2 expression, BCL2 translocation, and a MYD88 L265P mutation. Conversely, a diffuse CSF1R + B-cells pattern was associated with a favorable progression-free survival. Second, the histological expression of CSF1R was also correlated with 10 CSF1R-related markers including CSF1, STAT3, NFKB1, Ki67, MYC, PD-L1, TNFAIP8, IKAROS, CD163, and CD68. CSF1R moderately correlated with STAT3, TNFAIP8, CD68, and CD163 in the cases with the CSF1R + TAMs pattern. In addition, machine learning modeling predicted the CSF1R immunohistochemical expression with high accuracy using regression, generalized linear, an artificial intelligence neural network (multilayer perceptron), and support vector machine (SVM) analyses. Finally, a multilayer perceptron analysis predicted the genes associated with the CSF1R gene expression using the GEO GSE10846 DLBCL series of the Lymphoma/Leukemia Molecular Profiling Project (LLMPP), with correlation to the whole set of 20,683 genes as well as with an immuno-oncology cancer panel of 1790 genes. In addition, CSF1R positively correlated with SIRPA and inversely with CD47. In conclusion, the CSF1R histological pattern correlated with the progression-free survival of the patients of the Tokai series, and predictive analytics is a feasible strategy in DLBCL.
Collapse
|
37
|
Macrophages in multiple myeloma: key roles and therapeutic strategies. Cancer Metastasis Rev 2021; 40:273-284. [PMID: 33404860 DOI: 10.1007/s10555-020-09943-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022]
Abstract
Macrophages are a vital component of the tumour microenvironment and crucial mediators of tumour progression. In the last decade, significant strides have been made in understanding the crucial functional roles played by macrophages in the development of the plasma cell (PC) malignancy, multiple myeloma (MM). Whilst the interaction between MM PC and stromal cells within the bone marrow (BM) microenvironment has been extensively studied, we are only just starting to appreciate the multifaceted roles played by macrophages in disease progression. Accumulating evidence demonstrates that macrophage infiltration is associated with poor overall survival in MM. Indeed, macrophages influence numerous pathways critical for the initiation and progression of MM, including homing of malignant cells to BM, tumour cell growth and survival, drug resistance, angiogenesis and immune suppression. As such, therapeutic strategies aimed at targeting macrophages within the BM niche have promise in the clinical setting. This review will discuss the functions elicited by macrophages throughout different stages of MM and provide a comprehensive evaluation of potential macrophage-targeted therapies.
Collapse
|
38
|
Menzel L, Höpken UE, Rehm A. Angiogenesis in Lymph Nodes Is a Critical Regulator of Immune Response and Lymphoma Growth. Front Immunol 2020; 11:591741. [PMID: 33343570 PMCID: PMC7744479 DOI: 10.3389/fimmu.2020.591741] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor-induced remodeling of the microenvironment in lymph nodes (LNs) includes the formation of blood vessels, which goes beyond the regulation of metabolism, and shaping a survival niche for tumor cells. In contrast to solid tumors, which primarily rely on neo-angiogenesis, hematopoietic malignancies usually grow within pre-vascularized autochthonous niches in secondary lymphatic organs or the bone marrow. The mechanisms of vascular remodeling in expanding LNs during infection-induced responses have been studied in more detail; in contrast, insights into the conditions of lymphoma growth and lodging remain enigmatic. Based on previous murine studies and clinical trials in human, we conclude that there is not a universal LN-specific angiogenic program applicable. Instead, signaling pathways that are tightly connected to autochthonous and infiltrating cell types contribute variably to LN vascular expansion. Inflammation related angiogenesis within LNs relies on dendritic cell derived pro-inflammatory cytokines stimulating vascular endothelial growth factor-A (VEGF-A) expression in fibroblastic reticular cells, which in turn triggers vessel growth. In high-grade B cell lymphoma, angiogenesis correlates with poor prognosis. Lymphoma cells immigrate and grow in LNs and provide pro-angiogenic growth factors themselves. In contrast to infectious stimuli that impact on LN vasculature, they do not trigger the typical inflammatory and hypoxia-related stroma-remodeling cascade. Blood vessels in LNs are unique in selective recruitment of lymphocytes via high endothelial venules (HEVs). The dissemination routes of neoplastic lymphocytes are usually disease stage dependent. Early seeding via the blood stream requires the expression of the homeostatic chemokine receptor CCR7 and of L-selectin, both cooperate to facilitate transmigration of tumor and also of protective tumor-reactive lymphocytes via HEV structures. In this view, the HEV route is not only relevant for lymphoma cell homing, but also for a continuous immunosurveillance. We envision that HEV functional and structural alterations during lymphomagenesis are not only key to vascular remodeling, but also impact on tumor cell accessibility when targeted by T cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Lutz Menzel
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Uta E. Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Armin Rehm
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
39
|
Ruan J, Ouyang M, Zhang W, Luo Y, Zhou D. The effect of PD-1 expression on tumor-associated macrophage in T cell lymphoma. Clin Transl Oncol 2020; 23:1134-1141. [PMID: 33211280 DOI: 10.1007/s12094-020-02499-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/15/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Our study aimed to explore the programmed death 1 (PD-1) expression on tumor-associated macrophage (TAM) in T cell non-Hodgkin lymphoma (T-NHL) and its relationship with lymphoma prognosis. The effect of PD-1 expression on the function of macrophages was also studied. METHODS Multispectral image quantitative analysis was applied for detecting PD-1 expression on macrophages in T cell lymphoma tissues. The Kaplan-Meier analysis was performed to evaluate the value of PD-1 expression of TAM in predicting the overall survival of T-NHL. PD-1 overexpression THP-1-derived macrophage was constructed and was cocultured with Jurkat cells to explore the effect of PD-1 on macrophage function. RESULTS In 17 T cell lymphoma cases, the 1-year overall survival rate was significantly lower in patients with higher PD-1 expression on TAMs (0.25 vs 0.86, p < 0.05). After co-cultured with Jurkat cells, classically activated (M1)-related markers on PD-1 overexpressed macrophages were significantly lower than those on controls, while the expressions of alternatively activated (M2) related markers were similar. The PD-1 overexpressed macrophages showed inhibited phagocytosis (4.42% vs 40.7%, p < 0.001) and increased IL-10 secretion (144.48 pg/ml vs 32.32 pg/ml, p < 0.001). CONCLUSION High PD-1 expression on TAMs in T-NHL may predict poor prognosis. The PD-1 overexpression of macrophages significantly inhibited polarization of M1 macrophages and phagocytosis, and more IL-10 was excreted. These changes may enhance the pro-tumor effects of tumor microenvironment.
Collapse
Affiliation(s)
- J Ruan
- Department of Hematology, Chinese Academy of Medical Science, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - M Ouyang
- Department of Hematology, Chinese Academy of Medical Science, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.,Department of Cardiovascule, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - W Zhang
- Department of Hematology, Chinese Academy of Medical Science, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| | - Y Luo
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, China
| | - D Zhou
- Department of Hematology, Chinese Academy of Medical Science, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| |
Collapse
|
40
|
Chen J, Huang ZB, Liao CJ, Hu XW, Li SL, Qi M, Fan XG, Huang Y. LncRNA TP73-AS1/miR-539/MMP-8 axis modulates M2 macrophage polarization in hepatocellular carcinoma via TGF-β1 signaling. Cell Signal 2020; 75:109738. [DOI: 10.1016/j.cellsig.2020.109738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 01/30/2023]
|
41
|
Myers KV, Pienta KJ, Amend SR. Cancer Cells and M2 Macrophages: Cooperative Invasive Ecosystem Engineers. Cancer Control 2020; 27:1073274820911058. [PMID: 32129079 PMCID: PMC7066590 DOI: 10.1177/1073274820911058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Many aspects of cancer can be explained utilizing well-defined ecological
principles. Applying these principles to cancer, cancer cells are an invasive
species to a healthy organ ecosystem. In their capacity as ecosystem engineers,
cancer cells release cytokines that recruit monocytes to the tumor and polarize
them to M2-like protumor macrophages. Macrophages, recruited by the cancer
cells, act as a secondary invasive species. The ecosystem engineering functions
of M2-macrophages in turn support and stimulate cancer cell survival and
proliferation. The cooperative ecosystem engineering of both the primary
invasive species of the cancer cell and the secondary invasive species of the
M2-macrophage thus creates a vicious cycle of tumor promotion. Targeting a
specific aspect of this tumor-promoting ecosystem engineering, such as blocking
efferocytosis by M2-like macrophages, may improve the response to
standard-of-care anticancer therapies. This strategy has the potential to
redirect cooperative protumor ecosystem engineering toward an antitumor
ecosystem engineering strategy.
Collapse
Affiliation(s)
- Kayla V Myers
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins School of Medicine, Baltimore, MD, USA.,Department of Urology, The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kenneth J Pienta
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins School of Medicine, Baltimore, MD, USA.,Department of Urology, The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, MD, USA.,Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sarah R Amend
- Department of Urology, The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
42
|
Dossou AS, Sabnis N, Nagarajan B, Mathew E, Fudala R, Lacko AG. Lipoproteins and the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1272:93-116. [PMID: 32845504 DOI: 10.1007/978-3-030-48457-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
The tumor microenvironment (TME) plays a key role in enhancing the growth of malignant tumors and thus contributing to "aggressive phenotypes," supporting sustained tumor growth and metastasis. The precise interplay between the numerous components of the TME that contribute to the emergence of these aggressive phenotypes is yet to be elucidated and currently under intense investigation. The purpose of this article is to identify specific role(s) for lipoproteins as part of these processes that facilitate (or oppose) malignant growth as they interact with specific components of the TME during tumor development and treatment. Because of the scarcity of literature reports regarding the interaction of lipoproteins with the components of the tumor microenvironment, we were compelled to explore topics that were only tangentially related to this topic, to ensure that we have not missed any important concepts.
Collapse
Affiliation(s)
- Akpedje Serena Dossou
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Nirupama Sabnis
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Bhavani Nagarajan
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Ezek Mathew
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Rafal Fudala
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA.,Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Andras G Lacko
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA. .,Departments of Physiology/Anatomy and Pediatrics, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
43
|
Poreba M, Groborz KM, Rut W, Pore M, Snipas SJ, Vizovisek M, Turk B, Kuhn P, Drag M, Salvesen GS. Multiplexed Probing of Proteolytic Enzymes Using Mass Cytometry-Compatible Activity-Based Probes. J Am Chem Soc 2020; 142:16704-16715. [PMID: 32870676 PMCID: PMC7595764 DOI: 10.1021/jacs.0c06762] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The subset of the proteome that contains enzymes in their catalytically active form can be interrogated by using probes targeted toward individual specific enzymes. A subset of such enzymes are proteases that are frequently studied with activity-based probes, small inhibitors equipped with a detectable tag, commonly a fluorophore. Due to the spectral overlap of these commonly used fluorophores, multiplex analysis becomes limited. To overcome this, we developed a series of protease-selective lanthanide-labeled probes compatible with mass cytometry giving us the ability to monitor the activity of multiple proteases in parallel. Using these probes, we were able to identify the distribution of four proteases with different active site geometries in three cell lines and peripheral blood mononuclear cells. This provides a framework for the use of mass cytometry for multiplexed enzyme activity detection.
Collapse
Affiliation(s)
- Marcin Poreba
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Katarzyna M. Groborz
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Wioletta Rut
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Milind Pore
- University of Southern California, USC Michelson Center for Convergent Biosciences, Los Angeles, CA, USA
| | - Scott J. Snipas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Boris Turk
- Jozef Stefan Institute, Ljubljana, Slovenia
| | - Peter Kuhn
- University of Southern California, USC Michelson Center for Convergent Biosciences, Los Angeles, CA, USA
| | - Marcin Drag
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Guy S. Salvesen
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
44
|
Inflammatory Cells in Diffuse Large B Cell Lymphoma. J Clin Med 2020; 9:jcm9082418. [PMID: 32731512 PMCID: PMC7463675 DOI: 10.3390/jcm9082418] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/17/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL), known as the most common non-Hodgkin lymphoma (NHL) subtype, is characterized by high clinical and biological heterogeneity. The tumor microenvironment (TME), in which the tumor cells reside, is crucial in the regulation of tumor initiation, progression, and metastasis, but it also has profound effects on therapeutic efficacy. The role of immune cells during DLBCL development is complex and involves reciprocal interactions between tumor cells, adaptive and innate immune cells, their soluble mediators and structural components present in the tumor microenvironment. Different immune cells are recruited into the tumor microenvironment and exert distinct effects on tumor progression and therapeutic outcomes. In this review, we focused on the role of macrophages, Neutrophils, T cells, natural killer cells and dendritic cells in the DLBCL microenvironment and their implication as target for DLBCL treatment. These new therapies, carried out by the induction of adaptive immunity through vaccination or passive of immunologic effectors delivery, enhance the ability of the immune system to react against the tumor antigens inducing the destruction of tumor cells.
Collapse
|
45
|
Witschen PM, Chaffee TS, Brady NJ, Huggins DN, Knutson TP, LaRue RS, Munro SA, Tiegs L, McCarthy JB, Nelson AC, Schwertfeger KL. Tumor Cell Associated Hyaluronan-CD44 Signaling Promotes Pro-Tumor Inflammation in Breast Cancer. Cancers (Basel) 2020; 12:E1325. [PMID: 32455980 PMCID: PMC7281239 DOI: 10.3390/cancers12051325] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer has been conceptualized as a chronic wound with a predominance of tumor promoting inflammation. Given the accumulating evidence that the microenvironment supports tumor growth, we investigated hyaluronan (HA)-CD44 interactions within breast cancer cells, to determine whether this axis directly impacts the formation of an inflammatory microenvironment. Our results demonstrate that breast cancer cells synthesize and fragment HA and express CD44 on the cell surface. Using RNA sequencing approaches, we found that loss of CD44 in breast cancer cells altered the expression of cytokine-related genes. Specifically, we found that production of the chemokine CCL2 by breast cancer cells was significantly decreased after depletion of either CD44 or HA. In vivo, we found that CD44 deletion in breast cancer cells resulted in a delay in tumor formation and localized progression. This finding was accompanied by a decrease in infiltrating CD206+ macrophages, which are typically associated with tumor promoting functions. Importantly, our laboratory results were supported by human breast cancer patient data, where increased HAS2 expression was significantly associated with a tumor promoting inflammatory gene signature. Because high levels of HA deposition within many tumor types yields a poorer prognosis, our results emphasize that HA-CD44 interactions potentially have broad implications across multiple cancers.
Collapse
Affiliation(s)
- Patrice M. Witschen
- Comparative and Molecular Biosciences Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Thomas S. Chaffee
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (T.S.C.); (D.N.H.); (T.P.K.); (R.S.L.); (S.A.M.); (J.B.M.)
| | - Nicholas J. Brady
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Danielle N. Huggins
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (T.S.C.); (D.N.H.); (T.P.K.); (R.S.L.); (S.A.M.); (J.B.M.)
| | - Todd P. Knutson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (T.S.C.); (D.N.H.); (T.P.K.); (R.S.L.); (S.A.M.); (J.B.M.)
- University of Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rebecca S. LaRue
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (T.S.C.); (D.N.H.); (T.P.K.); (R.S.L.); (S.A.M.); (J.B.M.)
- University of Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sarah A. Munro
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (T.S.C.); (D.N.H.); (T.P.K.); (R.S.L.); (S.A.M.); (J.B.M.)
- University of Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lyubov Tiegs
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
| | - James B. McCarthy
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (T.S.C.); (D.N.H.); (T.P.K.); (R.S.L.); (S.A.M.); (J.B.M.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Andrew C. Nelson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (T.S.C.); (D.N.H.); (T.P.K.); (R.S.L.); (S.A.M.); (J.B.M.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Kathryn L. Schwertfeger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (T.S.C.); (D.N.H.); (T.P.K.); (R.S.L.); (S.A.M.); (J.B.M.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
46
|
Huang L, Wang Z, Chang Y, Wang K, Kang X, Huang R, Zhang Y, Chen J, Zeng F, Wu F, Zhao Z, Li G, Huang H, Jiang T, Hu H. EFEMP2 indicates assembly of M0 macrophage and more malignant phenotypes of glioma. Aging (Albany NY) 2020; 12:8397-8412. [PMID: 32396873 PMCID: PMC7244085 DOI: 10.18632/aging.103147] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 02/19/2020] [Indexed: 05/11/2023]
Abstract
Immune response mediated by macrophages is critical in tumor progression and implicates new targets in potential efficient immunotherapies. Tumor associated macrophages (TAM) are divided into either polarized M1 or M2 phenotype depending on different regulators of polarization and pro- or anti-oncogenic roles they play. Glioma-infiltrated TAMs have been newly reported contrary to the current polarization dogma. Instead, macrophages in glioma exhibit a continuum phenotype between the M1- and M2-like TAM that resembling M0 macrophage. Here we proposed an OS (overall survival)-correlated gene EFEMP2 (EGF containing fibulin-like extracellular matrix protein 2) via screening with transcriptional expression levels and methylation data in two glioma databases. EFEMP2 was found highly expressed in glioma of higher WHO grade and Mesenchymal subtype glioma, and its transcriptional level could predict OS efficiently in validation datasets. EFEMP2 exhibited a remarkable preference of intercellular expression. In vitro assay showed that EFEMP2's level in medium was closely related to glioma cells' growth. Moreover, EFEMP2 expression level was remarkably correlated with immunological responses. M0-like macrophage as a feature of malignancy of glioblastoma revealed distinct assembly in glioma with high level of EFEMP2. These results revealed EFEMP2's role as a potential characteristic marker of malignant glioma, which are enriched of M0 macrophage.
Collapse
Affiliation(s)
- Lijie Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Zheng Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Yuanhao Chang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Kuanyu Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Xun Kang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Ruoyu Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Ying Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Jing Chen
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Zheng Zhao
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Hua Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| |
Collapse
|
47
|
Han ZP, Liu DB, Wu LQ, Li Q, Wang ZG, Zang XF. IL-1β secreted by macrophage M2 promotes metastasis of osteosarcoma via NF-κB/miR-181α-5p/RASSF1A/Wnt pathway. Transl Cancer Res 2020; 9:2721-2733. [PMID: 35117631 PMCID: PMC8798966 DOI: 10.21037/tcr.2020.02.52] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/11/2020] [Indexed: 11/06/2022]
Abstract
BACKGROUND Ras-associated domain family protein1 isoform A (RASSF1A) was significantly absent in clinical samples and many osteosarcoma (OS) cell lines. Overexpression of RASSF1A could suppress OS metastasis, which may be mediated by tumor-associated macrophages polarized M2 (M2-TAMs). However, the relationship between IL-1β secreted by M2-TAMs and RASSF1A remains unknown. METHODS The expression levels of M2-TAMs markers CD68 and CD204 were measured by flow cytometry, and arginase-1 (Arg-1) and interleukin-1β (IL-1β) secreted by M2-TAMs were examined by real-time quantitative PCR (RT-qPCR). MTT assay was employed to determine the proliferation of OS cells, while scratch wound healing assay and Transwell assay were used to evaluate their migration and invasion, respectively. The level of miR-181α-5p was measured by RT-qPCR, while the levels of RASSF1A, GSK-3β, p-GSK-3β, β-catenin, MMP-2 and MMP-9 were evaluated by Western blot. The direct binding of miR-181α-5p and RASSF1A was identified using dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. RESULTS The levels of CD68, CD204, Arg-1 and IL-1β were elevated in M2-TAMs compared with control group. Overexpression of RASSF1A and knockdown of miR-181α-5p could both suppress invasion and migration of OS cells through Wnt pathway. IL-1β secreted by M2-TAMs facilitated the OS metastasis via RASSF1A/Wnt pathway, which could be targeted by miR-181α-5p and affected by nuclear factor-kappa B (NF-κB). CONCLUSIONS IL-1β secreted by M2-TAMs contributed to OS metastasis, which could be suppressed by knockdown of miR-181α-5p or overexpression of RASSF1A through NF-κB/miR-181α-5p/RASSF1A/Wnt pathway. These findings can guide new target discovery for drug development in OS treatment.
Collapse
Affiliation(s)
- Zhi-Peng Han
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Dong-Biao Liu
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Liu-Qing Wu
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Qin Li
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Zheng-Guang Wang
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Xiao-Fang Zang
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| |
Collapse
|
48
|
Amini MA, Abbasi AZ, Cai P, Lip H, Gordijo CR, Li J, Chen B, Zhang L, Rauth AM, Wu XY. Combining Tumor Microenvironment Modulating Nanoparticles with Doxorubicin to Enhance Chemotherapeutic Efficacy and Boost Antitumor Immunity. J Natl Cancer Inst 2020; 111:399-408. [PMID: 30239773 DOI: 10.1093/jnci/djy131] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 05/12/2018] [Accepted: 06/29/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tumor microenvironment (TME) and associated multiple factors are found to contribute to the failures in cancer therapies, including chemo- and immunotherapy. Here we report a new multimodal strategy that uses a bioreactive multifunctional hybrid polymer-lipid encapsulated manganese dioxide nanoparticle (PLMD NP) system to remodel the TME, suppress drug resistance factors, reverse immunosuppressive conditions, and enhance chemotherapy efficacy. METHODS The influence of PLMD NPs on enhancing cellular uptake in EMT6 mouse breast cancer cells and tumor penetration of doxorubicin (DOX) in EMT6 orthotopic breast tumor mouse model was evaluated using confocal microscopy (n = 3-4). Immunohistochemistry was employed to examine the effect of PLMD NPs on downregulating hypoxia-induced drug resistance proteins and anticancer activity of DOX (n = 3-4). The efficacy of the combination therapy with PLMD NPS and DOX was assessed in murine EMT6 (n = 15-23) and 4T1 (n = 7) orthotopic breast tumor mouse models. Rechallenge and splenocyte transfer were performed to validate the stimulation of adaptive tumor immunity in the surviving mice. RESULTS PLMD NPs enhanced intratumoral penetration and efficacy of DOX, and reduced intratumoral expression of P-glycoprotein, p53, and carbonic anhydrase IX by 74.5%, 38.0%, and 58.8% vs saline control, respectively. Combination treatment with PLMD NPs and DOX increased the number of tumor-infiltrated CD8+ T cells and resulted in up to 60.0% complete tumor regression. Of naïve mice (n = 7) that received splenocytes from the PLMD+DOX-treated surviving mice, 57.1% completely suppressed tumor growth whereas 100% of mice that received splenocytes from DOX-treated mice (n = 3) and the control group (n = 7) showed rapid tumor growth. CONCLUSIONS The clinically suitable PLMD NPs can effectively downregulate TME-associated drug resistance and immunosuppression. The combination therapy with PLMD NPs and DOX is a multimodal and translational treatment approach for enhancing chemotherapeutic efficacy and boosting antitumor immunity.
Collapse
Affiliation(s)
- Mohammad Ali Amini
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Azhar Z Abbasi
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Ping Cai
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - HoYin Lip
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Claudia R Gordijo
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Jason Li
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Branson Chen
- Department of Laboratory Medicine and Pathobiology and Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Li Zhang
- Toronto General Research Institute, The University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology and Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Andrew M Rauth
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
Li N, Liu C, Ma G, Tseng Y, Pan D, Chen J, Li F, Zeng X, Luo T, Chen S. Asparaginyl endopeptidase may promote liver sinusoidal endothelial cell angiogenesis via PI3K/Akt pathway. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2020; 111:214-222. [PMID: 30507245 DOI: 10.17235/reed.2018.5709/2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS pathological angiogenesis plays an important role in the progression of chronic liver diseases. Asparaginyl endopeptidase (AEP) participates in tumor angiogenesis and was recently shown to be associated with liver fibrosis. This study aimed to explore the effect of AEP on liver sinusoidal endothelial cell (LSECs) angiogenesis and determine the underlying mechanism. METHODS cultured LSECs were infected with lentiviruses in order to suppress AEP expression (AEP-KD1, AEP-KD2). The effect of AEP on LSECs proliferation, apoptosis and migration were subsequently determined by a CCK8 assay, flow cytometry and wound-healing and Transwell assays, respectively, in AEP knocked-down and control LSECs. The expression of the endothelial cell surface markers CD31, CD34 and von Willebrand factor (vWF) were detected by immunofluorescence assay and western blot. The angiogenic factors, vascular endothelial growth factor receptor 2 (VEGFR2) and interleukin 8 (IL 8) were detected by real-time PCR and western blot. The effect of AEP on vessel tube formation by LSECs was examined by Matrigel™ tube-formation assay. Phosphoinositide 3-kinase (PI3K)/Akt expression and phosphorylation were detected by western blot. RESULTS AEP was effectively knocked down by lentivirus infection in LSECs. Down-regulation of AEP expression significantly decreased proliferation and migration and increased apoptosis of LSECs. Moreover, expression levels of the endothelial cell surface markers CD31, CD34 and vWF, as well as angiogenic factors VEGFR2 and IL 8, were also reduced after AEP was knocked-down. The vessel tube formation abilities of AEP-KD1 and AEP-KD2 LSECs were significantly inhibited compared with LSECs without AEP knocked-down. Down-regulation of AEP also inhibited the phosphorylation of PI3K and Akt. CONCLUSION AEP promotes LSECs angiogenesis in vitro, possibly via the PI3K/Akt pathway. AEP may therefore be a potential therapeutic target for preventing the progression of liver fibrosis.
Collapse
Affiliation(s)
- Na Li
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Chu Liu
- Department of Gastroenterology, Zhongshan Hospital, Fudan Ubiversity, China
| | - Guifen Ma
- Department of Radiotherapy, Zhongshan Hospital, Fudan University, China
| | - Yujen Tseng
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Duyi Pan
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Jie Chen
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Feng Li
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Xiaoqing Zeng
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Tiancheng Luo
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Shiyao Chen
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| |
Collapse
|
50
|
Roussel M, Lhomme F, Roe CE, Bartkowiak T, Gravelle P, Laurent C, Fest T, Irish JM. Mass cytometry defines distinct immune profile in germinal center B-cell lymphomas. Cancer Immunol Immunother 2020; 69:407-420. [PMID: 31919622 PMCID: PMC7764565 DOI: 10.1007/s00262-019-02464-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/26/2019] [Indexed: 02/08/2023]
Abstract
Tumor-associated macrophage and T-cell subsets are implicated in the pathogenesis of diffuse large B-cell lymphoma, follicular lymphoma, and classical Hodgkin lymphoma. Macrophages provide essential mechanisms of tumor immune evasion through checkpoint ligand expression and secretion of suppressive cytokines. However, normal and tumor-associated macrophage phenotypes are less well characterized than those of tumor-infiltrating T-cell subsets, and it would be especially valuable to know whether the polarization state of macrophages differs across lymphoma tumor microenvironments. Here, an established mass cytometry panel designed to characterize myeloid-derived suppressor cells and known macrophage maturation and polarization states was applied to characterize B-lymphoma tumors and non-malignant human tissue. High-dimensional single-cell analyses were performed using dimensionality reduction and clustering tools. Phenotypically distinct intra-tumor macrophage subsets were identified based on abnormal marker expression profiles that were associated with lymphoma tumor types. While it had been proposed that measurement of CD163 and CD68 might be sufficient to reveal macrophage subsets in tumors, results here indicated that S100A9, CCR2, CD36, Slan, and CD32 should also be measured to effectively characterize lymphoma-specific tumor macrophages. Additionally, the presence of phenotypically distinct, abnormal macrophage populations was closely linked to the phenotype of intra-tumor T-cell populations, including PD-1 expressing T cells. These results further support the close links between macrophage polarization and T-cell functional state, as well as the rationale for targeting tumor-associated macrophages in cancer immunotherapies.
Collapse
Affiliation(s)
- Mikael Roussel
- Laboratoire Hématologie, CHU Pontchaillou, Centre Hospitalier Universitaire de Rennes, Pôle Biologie, 2 rue Henri Le Guilloux, 35033, Rennes, France.
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France.
| | - Faustine Lhomme
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France
| | - Caroline E Roe
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 740B Preston Building, 2220 Pierce Avenue, Nashville, TN, 37232-6840, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Todd Bartkowiak
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 740B Preston Building, 2220 Pierce Avenue, Nashville, TN, 37232-6840, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Pauline Gravelle
- Service Anatomie et Cytologie Pathologiques and UMR1037, Toulouse, France
| | - Camille Laurent
- Service Anatomie et Cytologie Pathologiques and UMR1037, Toulouse, France
| | - Thierry Fest
- Laboratoire Hématologie, CHU Pontchaillou, Centre Hospitalier Universitaire de Rennes, Pôle Biologie, 2 rue Henri Le Guilloux, 35033, Rennes, France
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 740B Preston Building, 2220 Pierce Avenue, Nashville, TN, 37232-6840, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|